Browse
Search for other papers by Kreepa G Kooblall in
Google Scholar
PubMed
Search for other papers by Victoria J Stokes in
Google Scholar
PubMed
Search for other papers by Omair A Shariq in
Google Scholar
PubMed
Search for other papers by Katherine A English in
Google Scholar
PubMed
Search for other papers by Mark Stevenson in
Google Scholar
PubMed
Search for other papers by John Broxholme in
Google Scholar
PubMed
Search for other papers by Benjamin Wright in
Google Scholar
PubMed
Search for other papers by Helen E Lockstone in
Google Scholar
PubMed
Search for other papers by David Buck in
Google Scholar
PubMed
Search for other papers by Simona Grozinsky-Glasberg in
Google Scholar
PubMed
Search for other papers by Christopher J Yates in
Google Scholar
PubMed
Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
Search for other papers by Rajesh V Thakker in
Google Scholar
PubMed
Search for other papers by Kate E Lines in
Google Scholar
PubMed
Multiple endocrine neoplasia type 1 (MEN1), caused by mutations in the MEN1 gene encoding menin, is an autosomal dominant disorder characterised by the combined occurrence of parathyroid, pituitary and pancreatic neuroendocrine tumours (NETs). Development of these tumours is associated with wide variations in their severity, order and ages (from <5 to >80 years), requiring life-long screening. To improve tumour surveillance and quality of life, better circulating biomarkers, particularly for pancreatic NETs that are associated with higher mortality, are required. We, therefore, examined the expression of circulating miRNA in the serum of MEN1 patients. Initial profiling analysis followed by qRT-PCR validation studies identified miR-3156-5p to be significantly downregulated (−1.3 to 5.8-fold, P < 0.05–0.0005) in nine MEN1 patients, compared to matched unaffected relatives. MEN1 knock-down experiments in BON-1 human pancreatic NET cells resulted in reduced MEN1 (49%, P < 0.05), menin (54%, P < 0.05) and miR-3156-5p expression (20%, P < 0.005), compared to control-treated cells, suggesting that miR-3156-5p downregulation is a consequence of loss of MEN1 expression. In silico analysis identified mortality factor 4-like 2 (MOR4FL2) as a potential target of miR-3156-5p, and in vitro functional studies in BON-1 cells transfected with either miR-3156-5p mimic or inhibitors showed that the miR-3156-5p mimic significantly reduced MORF4L2 protein expression (46%, P < 0.005), while miR-3156-5p inhibitor significantly increased MORF4L2 expression (1.5-fold, P < 0.05), compared to control-treated cells, thereby confirming that miR-3156-5p regulates MORF4L2 expression. Thus, the inverse relationship between miR-3156-5p and MORF4L2 expression represents a potential serum biomarker that could facilitate the detection of NET occurrence in MEN1 patients.
Regional Centre for Endocrinology and Diabetes, Royal Victoria Hospital, Belfast Health & Social Care Trust, Belfast, UK
Search for other papers by Paul Benjamin Loughrey in
Google Scholar
PubMed
Search for other papers by Federico Roncaroli in
Google Scholar
PubMed
Search for other papers by Estelle Healy in
Google Scholar
PubMed
Search for other papers by Philip Weir in
Google Scholar
PubMed
Search for other papers by Madhu Basetti in
Google Scholar
PubMed
Search for other papers by Ruth T Casey in
Google Scholar
PubMed
Search for other papers by Steven J Hunter in
Google Scholar
PubMed
Search for other papers by Márta Korbonits in
Google Scholar
PubMed
Pituitary neuroendocrine tumours (PitNETs) associated with paragangliomas or phaeochromocytomas are rare. SDHx variants are estimated to be associated with 0.3–1.8% of PitNETs. Only a few case reports have documented the association with MAX variants. Prolactinomas are the most common PitNETs occurring in patients with SDHx variants, followed by somatotrophinomas, clinically non-functioning tumours and corticotrophinomas. One pituitary carcinoma has been described. SDHC, SDHB and SDHA mutations are inherited in an autosomal dominant fashion and tumorigenesis seems to adhere to Knudson’s two-hit hypothesis. SDHD and SDHAF2 mutations most commonly have paternal inheritance. Immunohistochemistry for SDHB or MAX and loss of heterozygosity analysis can support the assessment of pathogenicity of the variants. Metabolomics is promising in the diagnosis of SDHx-related disease. Future research should aim to further clarify the role of SDHx and MAX variants or other genes in the molecular pathogenesis of PitNETs, including pseudohypoxic and kinase signalling pathways along with elucidating epigenetic mechanisms to predict tumour behaviour.
The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
Search for other papers by Anastasia Alataki in
Google Scholar
PubMed
The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
Search for other papers by Mitch Dowsett in
Google Scholar
PubMed
Endocrine therapies are the main treatment strategies for the clinical management of hormone-dependent breast cancer. Despite prolonged time to recurrence in the adjuvant setting and the initial clinical responses in the metastatic setting, many patients eventually encounter tumour relapse due to acquired resistance to these agents. Other patients experience a lack of tumour regression at the beginning of treatment indicating de novo resistance that significantly limits its efficacy in the clinic. There is compelling evidence that human epidermal growth factor receptor-2 (HER2) overexpression contributes to resistance to endocrine therapies in oestrogen receptor-positive (ER+) breast cancer. ER+/HER2+ tumours comprise about 10% of all breast cancer cases and about 60% of the whole set of HER2+ tumours. Most patients with primary ER+/HER2+ disease will receive antibody-based HER2-targeted therapy, but this is generally for no more than one year while endocrine treatment is usually for at least 5 years. A number of HER2-kinase inhibitors are also now in clinical use or in clinical trials, and the interaction of these with endocrine treatment may differ from that of antibody treatment. In this review article, we aim to summarise knowledge on molecular mechanisms of breast cancer resistance to endocrine therapies attributable to the impact of HER2 signalling on endocrine sensitivity, to discuss data from clinical trials addressing the role of HER2 in the development of endocrine resistance in the metastatic, neoadjuvant and adjuvant settings and to explore rational new therapeutic strategies.
Search for other papers by Adriana Albani in
Google Scholar
PubMed
Search for other papers by Luis Gustavo Perez-Rivas in
Google Scholar
PubMed
Search for other papers by Sicheng Tang in
Google Scholar
PubMed
Search for other papers by Julia Simon in
Google Scholar
PubMed
Search for other papers by Kristin Elisabeth Lucia in
Google Scholar
PubMed
Search for other papers by Paula Colón-Bolea in
Google Scholar
PubMed
Search for other papers by Jochen Schopohl in
Google Scholar
PubMed
Search for other papers by Sigrun Roeber in
Google Scholar
PubMed
Search for other papers by Michael Buchfelder in
Google Scholar
PubMed
Search for other papers by Roman Rotermund in
Google Scholar
PubMed
Search for other papers by Jörg Flitsch in
Google Scholar
PubMed
Search for other papers by Jun Thorsteinsdottir in
Google Scholar
PubMed
Search for other papers by Jochen Herms in
Google Scholar
PubMed
Medicover Neuroendocrinology, Munich, Germany
Search for other papers by Günter Stalla in
Google Scholar
PubMed
Search for other papers by Martin Reincke in
Google Scholar
PubMed
Search for other papers by Marily Theodoropoulou in
Google Scholar
PubMed
Cushing’s disease is a rare but devastating and difficult to manage condition. The somatostatin analogue pasireotide is the only pituitary-targeting pharmaceutical approved for the treatment of Cushing’s disease but is accompanied by varying efficacy and potentially severe side effects. Finding means to predict which patients are more likely to benefit from this treatment may improve their management. More than half of corticotroph tumours harbour mutations in the USP8 gene, and there is evidence of higher somatostatin receptor 5 (SSTR5) expression in the USP8-mutant tumours. Pasireotide has a high affinity for SSTR5, indicating that these tumours may be more sensitive to treatment. To test this hypothesis, we examined the inhibitory action of pasireotide on adrenocorticotrophic hormone synthesis in primary cultures of human corticotroph tumour with assessed USP8 mutational status and in immortalized murine corticotroph tumour cells overexpressing human USP8 mutants frequent in Cushing’s disease. Our in vitro results demonstrate that pasireotide exerts a higher antisecretory response in USP8-mutant corticotroph tumours. Overexpressing USP8 mutants in a murine corticotroph tumour cell model increased endogenous somatostatin receptor 5 (Sstr5) transcription. The murine Sstr5 promoter has two binding sites for the activating protein 1 (AP-1) and USP8 mutants possibly to mediate their action by stimulating AP-1 transcriptional activity. Our data corroborate the USP8 mutational status as a potential marker of pasireotide response and describe a potential mechanism through which USP8 mutants may regulate SSTR5 gene expression.
Search for other papers by Anna Angelousi in
Google Scholar
PubMed
Search for other papers by Aimee R Hayes in
Google Scholar
PubMed
Search for other papers by Eleftherios Chatzellis in
Google Scholar
PubMed
Search for other papers by Gregory A Kaltsas in
Google Scholar
PubMed
Green Templeton College, University of Oxford, Oxford, UK
Centre for Endocrinology, Barts and the London School of Medicine, London, UK
Search for other papers by Ashley B Grossman in
Google Scholar
PubMed
Medullary thyroid carcinoma (MTC) is a rare malignancy comprising 1–2% of all thyroid cancers in the United States. Approximately 20% of cases are familial, secondary to a germline RET mutation, while the remaining 80% are sporadic and also harbour a somatic RET mutation in more than half of all cases. Up to 15–20% of patients will present with distant metastatic disease, and retrospective series report a 10-year survival of 10–40% from time of first metastasis. Historically, systemic therapies for metastatic MTC have been limited, and cytotoxic chemotherapy has demonstrated poor objective response rates. However, in the last decade, targeted therapies, particularly multitargeted tyrosine kinase inhibitors (TKIs), have demonstrated prolonged progression-free survival in advanced and progressive MTC. Both cabozantinib and vandetanib have been approved as first-line treatment options in many countries; nevertheless, their use is limited by high toxicity rates and dose reductions are often necessary. New generation TKIs, such as selpercatinib or pralsetinib, that exhibit selective activity against RET, have recently been approved as a second-line treatment option, and they exhibit a more favourable side-effect profile. Peptide receptor radionuclide therapy or immune checkpoint inhibitors may also constitute potential therapeutic options in specific clinical settings. In this review, we aim to present all current therapeutic options available for patients with progressive MTC, as well as new or as yet experimental treatments.
Search for other papers by William Beimers in
Google Scholar
PubMed
Search for other papers by Megan Braun in
Google Scholar
PubMed
Search for other papers by Kaleb Schwinefus in
Google Scholar
PubMed
Search for other papers by Keenan Pearson in
Google Scholar
PubMed
Search for other papers by Brandon Wilbanks in
Google Scholar
PubMed
Search for other papers by Louis James Maher in
Google Scholar
PubMed
A fascinating class of familial paraganglioma (PGL) neuroendocrine tumors is driven by the loss of the tricarboxylic acid (TCA) cycle enzyme succinate dehydrogenase (SDH) resulting in succinate accumulation as an oncometabolite and other metabolic derangements. Here, we exploit a Saccharomyces cerevisiae yeast model of SDH loss where accumulating succinate, and possibly reactive oxygen species, poison a dioxygenase enzyme required for sulfur scavenging. Using this model, we performed a chemical suppression screen for compounds that relieve dioxygenase inhibition. After testing 1280 pharmaceutically active compounds, we identified meclofenoxate HCl and its hydrolysis product, dimethylaminoethanol (DMAE), as suppressors of dioxygenase intoxication in SDH-loss yeast cells. We show that DMAE acts to alter metabolism so as to normalize the succinate:2-ketoglutarate ratio, improving dioxygenase function. This study raises the possibility that oncometabolite effects might be therapeutically suppressed by drugs that rewire metabolism to reduce the flux of carbon into pathological metabolic pathways.
Search for other papers by Susan Richter in
Google Scholar
PubMed
Search for other papers by Bei Qiu in
Google Scholar
PubMed
Search for other papers by Mirthe Ghering in
Google Scholar
PubMed
Search for other papers by Carola Kunath in
Google Scholar
PubMed
Search for other papers by Georgiana Constantinescu in
Google Scholar
PubMed
Search for other papers by Charlotte Luths in
Google Scholar
PubMed
Search for other papers by Christina Pamporaki in
Google Scholar
PubMed
Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
Search for other papers by Nicole Bechmann in
Google Scholar
PubMed
Search for other papers by Leah Meuter in
Google Scholar
PubMed
Search for other papers by Aleksandra Kwapiszewska in
Google Scholar
PubMed
Medicover Oldenburg MVZ, Oldenburg, Germany
Search for other papers by Timo Deutschbein in
Google Scholar
PubMed
Department for Endocrinology, Diabetology and Clinical Nutrition, UniversitätsSpital Zürich, Zurich, Switzerland
Search for other papers by Svenja Nölting in
Google Scholar
PubMed
Search for other papers by Mirko Peitzsch in
Google Scholar
PubMed
Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
Search for other papers by Mercedes Robledo in
Google Scholar
PubMed
Search for other papers by Aleksander Prejbisz in
Google Scholar
PubMed
Search for other papers by Karel Pacak in
Google Scholar
PubMed
Klinik für Hals-Nasen-Ohrenheilkunde, Kopf- und Hals-Chirurgie, Plastische Operationen, Städtisches Klinikum Dresden, Akademisches Lehrkrankenhaus der Technischen Universität Dresden, Dresden, Germany
Search for other papers by Volker Gudziol in
Google Scholar
PubMed
Search for other papers by Henri J L M Timmers in
Google Scholar
PubMed
Department of Medicine III, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
Search for other papers by Graeme Eisenhofer in
Google Scholar
PubMed
Head and neck paragangliomas (HNPGLs) are tumors of parasympathetic origin that occur at variable locations and are often secondary to germline mutations in succinate dehydrogenase (SDH) subunit genes. Occasionally, these tumors produce catecholamines. Here, we assessed whether different locations of HNPGLs relate to the presence of SDHx mutations, catecholamine production and other presentations. In this multicenter study, we collected clinical and biochemical data from 244 patients with HNPGLs and 71 patients without HNPGLs. We clarified that jugulotympanic HNPGLs have distinct features. In particular, 88% of jugulotympanic HNPGLs arose in women, among whom only 24% occurred due to SDHx mutations compared to 55% in men. Jugulotympanic HNPGLs were also rarely bilateral, were of a smaller size and were less often metastatic compared to carotid body and vagal HNPGLs. Furthermore, we showed that plasma concentrations of methoxytyramine (MTY) were higher (P < 0.0001) in patients with HNPGL than without HNPGL, whereas plasma normetanephrine did not differ. Only 3.7% of patients showed strong increases in plasma normetanephrine. Plasma MTY was positively related to tumor size but did not relate to the presence of SDHx mutations or tumor location. Our findings confirm that increases in plasma MTY represent the main catecholamine-related biochemical feature of patients with HNPGLs. We expect that more sensitive analytical methods will make biochemical testing of HNPGLs more practical in the future and enable more than the current 30% of patients to be identified with dopamine-producing HNPGLs. The sex-dependent differences in the development of HNPGLs may have relevance to the diagnosis, management and outcomes of these tumors.
Dan L. Duncan Comprehensive Cancer Center, Houston, Texas, USA
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Salma Kaochar in
Google Scholar
PubMed
Search for other papers by Aleksandra Rusin in
Google Scholar
PubMed
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Christopher Foley in
Google Scholar
PubMed
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Kimal Rajapakshe in
Google Scholar
PubMed
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Matthew Robertson in
Google Scholar
PubMed
Search for other papers by Darlene Skapura in
Google Scholar
PubMed
Search for other papers by Cammy Mason in
Google Scholar
PubMed
Search for other papers by Karen Berman De Ruiz in
Google Scholar
PubMed
Search for other papers by Alexey Mikhailovich Tyryshkin in
Google Scholar
PubMed
Search for other papers by Jenny Deng in
Google Scholar
PubMed
Search for other papers by Jin Na Shin in
Google Scholar
PubMed
Search for other papers by Warren Fiskus in
Google Scholar
PubMed
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Jianrong Dong in
Google Scholar
PubMed
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Department of Education, Innovation, and Technology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Shixia Huang in
Google Scholar
PubMed
Search for other papers by Nora M Navone in
Google Scholar
PubMed
Search for other papers by Christel M Davis in
Google Scholar
PubMed
Search for other papers by Erik A Ehli in
Google Scholar
PubMed
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Cristian Coarfa in
Google Scholar
PubMed
Dan L. Duncan Comprehensive Cancer Center, Houston, Texas, USA
Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
Search for other papers by Nicholas Mitsiades in
Google Scholar
PubMed
Castration-resistant prostate cancer (CRPC) remains highly lethal and in need of novel, actionable therapeutic targets. The pioneer factor GATA2 is a significant prostate cancer (PC) driver and is linked to poor prognosis. GATA2 directly promotes androgen receptor (AR) gene expression (both full-length and splice-variant) and facilitates AR binding to chromatin, recruitment of coregulators, and target gene transcription. Unfortunately, there is no clinically applicable GATA2 inhibitor available at the moment. Using a bioinformatics algorithm, we screened in silico 2650 clinically relevant drugs for a potential GATA2 inhibitor. Validation studies used cytotoxicity and proliferation assays, global gene expression analysis, RT-qPCR, reporter assay, reverse phase protein array analysis (RPPA), and immunoblotting. We examined target engagement via cellular thermal shift assay (CETSA), ChIP-qPCR, and GATA2 DNA-binding assay. We identified the vasodilator dilazep as a potential GATA2 inhibitor and confirmed on-target activity via CETSA. Dilazep exerted anticancer activity across a broad panel of GATA2-dependent PC cell lines in vitro and in a PDX model in vivo. Dilazep inhibited GATA2 recruitment to chromatin and suppressed the cell-cycle program, transcriptional programs driven by GATA2, AR, and c-MYC, and the expression of several oncogenic drivers, including AR, c-MYC, FOXM1, CENPF, EZH2, UBE2C, and RRM2, as well as of several mediators of metastasis, DNA damage repair, and stemness. In conclusion, we provide, via an extensive compendium of methodologies, proof-of-principle that a small molecule can inhibit GATA2 function and suppress its downstream AR, c-MYC, and other PC-driving effectors. We propose GATA2 as a therapeutic target in CRPC.
Department of Oncology, Haukeland University Hospital, Bergen, Norway
Search for other papers by Andreas Venizelos in
Google Scholar
PubMed
Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
Search for other papers by Hege Elvebakken in
Google Scholar
PubMed
Search for other papers by Aurel Perren in
Google Scholar
PubMed
Department of Oncology, Haukeland University Hospital, Bergen, Norway
Search for other papers by Oleksii Nikolaienko in
Google Scholar
PubMed
Department of Oncology, Haukeland University Hospital, Bergen, Norway
Search for other papers by Wei Deng in
Google Scholar
PubMed
Search for other papers by Inger Marie B Lothe in
Google Scholar
PubMed
Search for other papers by Anne Couvelard in
Google Scholar
PubMed
Search for other papers by Geir Olav Hjortland in
Google Scholar
PubMed
Department of Medical Radiation Physics, Lund University, Lund, Sweden
Search for other papers by Anna Sundlöv in
Google Scholar
PubMed
Search for other papers by Johanna Svensson in
Google Scholar
PubMed
Search for other papers by Harrish Garresori in
Google Scholar
PubMed
Search for other papers by Christian Kersten in
Google Scholar
PubMed
Department of Oncology, St.Olavs Hospital, Trondheim, Norway
Search for other papers by Eva Hofsli in
Google Scholar
PubMed
Department of Clinical Medicine, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
Search for other papers by Sönke Detlefsen in
Google Scholar
PubMed
Search for other papers by Merete Krogh in
Google Scholar
PubMed
Department of Clinical Science, University of Bergen, Bergen, Norway
Search for other papers by Halfdan Sorbye in
Google Scholar
PubMed
Department of Oncology, Haukeland University Hospital, Bergen, Norway
Search for other papers by Stian Knappskog in
Google Scholar
PubMed
High-grade (HG) gastroenteropancreatic (GEP) neuroendocrine neoplasms (NEN) are rare but have a very poor prognosis and represent a severely understudied class of tumours. Molecular data for HG GEP-NEN are limited, and treatment strategies for the carcinoma subgroup (HG GEP-NEC) are extrapolated from small-cell lung cancer (SCLC). After pathological re-evaluation, we analysed DNA from tumours and matched blood samples from 181 HG GEP-NEN patients; 152 neuroendocrine carcinomas (NEC) and 29 neuroendocrine tumours (NET G3). Based on the sequencing of 360 cancer-related genes, we assessed mutations and copy number alterations (CNA). For NEC, frequently mutated genes were TP53 (64%), APC (28%), KRAS (22%) and BRAF (20%). RB1 was only mutated in 14%, but CNAs affecting RB1 were seen in 34%. Other frequent copy number losses were ARID1A (35%), ESR1 (25%) and ATM (31%). Frequent amplifications/gains were found in MYC (51%) and KDM5A (45%). While these molecular features had limited similarities with SCLC, we found potentially targetable alterations in 66% of the NEC samples. Mutations and CNA varied according to primary tumour site with BRAF mutations mainly seen in colon (49%), and FBXW7 mutations mainly seen in rectal cancers (25%). Eight out of 152 (5.3%) NEC were microsatellite instable (MSI). NET G3 had frequent mutations in MEN1 (21%), ATRX (17%), DAXX, SETD2 and TP53 (each 14%). We show molecular differences in HG GEP-NEN, related to morphological differentiation and site of origin. Limited similarities to SCLC and a high fraction of targetable alterations indicate a high potential for better-personalized treatments.
Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
Search for other papers by Luise Eckardt in
Google Scholar
PubMed
Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
Search for other papers by Maria Prange-Barczynska in
Google Scholar
PubMed
The Department of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
Search for other papers by Emma J Hodson in
Google Scholar
PubMed
Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
Search for other papers by James W Fielding in
Google Scholar
PubMed
Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
Search for other papers by Xiaotong Cheng in
Google Scholar
PubMed
Search for other papers by Joanna D C C Lima in
Google Scholar
PubMed
Search for other papers by Samvid Kurlekar in
Google Scholar
PubMed
Search for other papers by Gillian Douglas in
Google Scholar
PubMed
Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
The Francis Crick Institute, London, UK
Search for other papers by Peter J Ratcliffe in
Google Scholar
PubMed
Search for other papers by Tammie Bishop in
Google Scholar
PubMed
Despite a general role for the HIF hydroxylase system in cellular oxygen sensing and tumour hypoxia, cancer-associated mutations of genes in this pathway, including PHD2, PHD1, EPAS1 (encoding HIF-2α) are highly tissue-restricted, being observed in pseudohypoxic pheochromocytoma and paraganglioma (PPGL) but rarely, if ever, in other tumours. In an effort to understand that paradox and gain insights into the pathogenesis of pseudohypoxic PPGL, we constructed mice in which the principal HIF prolyl hydroxylase, Phd2, is inactivated in the adrenal medulla using TH-restricted Cre recombinase. Investigation of these animals revealed a gene expression pattern closely mimicking that of pseudohypoxic PPGL. Spatially resolved analyses demonstrated a binary distribution of two contrasting patterns of gene expression among adrenal medullary cells. Phd2 inactivation resulted in a marked shift in this distribution towards a Pnmt −/Hif-2α +/Rgs5 + population. This was associated with morphological abnormalities of adrenal development, including ectopic TH+ cells within the adrenal cortex and external to the adrenal gland. These changes were ablated by combined inactivation of Phd2 with Hif-2α, but not Hif-1α. However, they could not be reproduced by inactivation of Phd2 in adult life, suggesting that they arise from dysregulation of this pathway during adrenal development. Together with the clinical observation that pseudohypoxic PPGL manifests remarkably high heritability, our findings suggest that this type of tumour likely arises from dysregulation of a tissue-restricted action of the PHD2/HIF-2α pathway affecting adrenal development in early life and provides a model for the study of the relevant processes.