1
|
Tatachar S, Rajan R, Hesaraghatta Shyamsunder A, Kapoor N. Obesity in patients with craniopharyngioma in the South Asian region - A distinct phenotype. OBESITY PILLARS 2024; 12:100139. [PMID: 39431051 PMCID: PMC11489328 DOI: 10.1016/j.obpill.2024.100139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024]
Abstract
Background Craniopharyngiomas are rare benign tumors located in the sellar and suprasellar region, with an incidence of 0.5-2 cases per million as reported in Western studies. Post-treatment, including surgery and/or radiotherapy, many patients develop significant obesity, primarily due to hypothalamic damage and associated complications such as hypopituitarism. In the South Asian population, genetic predisposition to obesity at lower BMIs, coupled with a carbohydrate-rich diet, may exacerbate obesity in craniopharyngioma patients, presenting a unique challenge. Methods This submission is a commentary based on a comprehensive literature review. The authors conducted the review using PubMed to focus on English-language articles covering hypothalamic obesity, craniopharyngioma and obesity in the South Asian population from 1939 to the present. Results The literature review revealed that 50-60 % of patients treated for craniopharyngioma develop obesity, predominantly linked to hypothalamic damage, although these data are mainly derived from Western studies. Hypopituitarism was frequently observed, further contributing to the obesity. Despite a caloric intake appropriate for the age and gender, these patients exhibited reduced physical activity as measured by wrist accelerometers. Patients with hypothalamic obesity due to craniopharyngioma are at risk for metabolic syndrome and cardiovascular morbidity. Additionally, visual impairment was common, leading to a decreased quality of life. The South Asian population, genetically predisposed to visceral obesity and a carbohydrate-rich diet, may display a distinct phenotype. Although multiple treatment modalities have been tried, there is no definite treatment modality available to counteract this condition at present. Conclusion South Asian phenotype of craniopharyngioma-related obesity is characterized by significant metabolic and hormonal dysregulation, influenced by both dietary and genetic factors. Nevertheless, there may be a lot to be still understood about this devastating, rapid, relentless hypothalamic obesity syndrome. Also, a higher morbidity rate within this population, underscores the need for further research to develop targeted interventions.
Collapse
Affiliation(s)
- Sreevatsa Tatachar
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, 632004, Vellore, Tamilnadu, India
| | - Remya Rajan
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, 632004, Vellore, Tamilnadu, India
| | | | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College, 632004, Vellore, Tamilnadu, India
| |
Collapse
|
2
|
Svendstrup M, Rasmussen AK, Kistorp C, Klose M, Andreassen M. Semaglutide treatment of hypothalamic obesity - a real-life data study. Pituitary 2024; 27:685-692. [PMID: 39120810 PMCID: PMC11513754 DOI: 10.1007/s11102-024-01429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE Patients with tumors involving the hypothalamic region are at high risk of developing morbid obesity due to disturbances in the appetite regulative nuclei in hypothalamus. We evaluated the effect of the Glucagon-like peptide 1 (GLP-1) analogue semaglutide in patients with hypothalamic obesity. METHODS We recorded weight changes from real-time data before and after treatment with semaglutide in patients with hypothalamic obesity from our outpatient clinic at the Department of Endocrinology at Rigshospitalet, from September 2020 to November 2023. RESULTS A total of 26 patients were included in this study (15 females, median age at initiation of semaglutide was 52 (range 18-65) years). Body mass index (BMI) at initial diagnosis was median 25 (range 20-38) kg/m2 while BMI at initiation of semaglutide was median 38 (range 28-58) kg/m2. All but one patient lost weight during semaglutide treatment with a mean weight loss of 13.4 kg (95% CI 10.3-16.5 kg, p = < 0.001) after 12 months corresponding to a loss in BMI of 4.4 kg/m2 (95% CI 3.4-5.4 kg/m2, p = < 0.001) with a median dosage of semaglutide of 1.6 (range 0.5-2.5) mg. Fifteen patients (58%) lost more than 10% and two patients (8%) lost more than 20% of initial body weight, respectively. CONCLUSION Treatment with semaglutide shows promising results in reducing body weight in patients with acquired hypothalamic obesity. Whether the weight reduction remains stable after long time follow-up needs further investigation.
Collapse
Affiliation(s)
- Mathilde Svendstrup
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Aase Krogh Rasmussen
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Caroline Kistorp
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marianne Klose
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mikkel Andreassen
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
3
|
Gan HW, Cerbone M, Dattani MT. Appetite- and Weight-Regulating Neuroendocrine Circuitry in Hypothalamic Obesity. Endocr Rev 2024; 45:309-342. [PMID: 38019584 PMCID: PMC11074800 DOI: 10.1210/endrev/bnad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 11/30/2023]
Abstract
Since hypothalamic obesity (HyOb) was first described over 120 years ago by Joseph Babinski and Alfred Fröhlich, advances in molecular genetic laboratory techniques have allowed us to elucidate various components of the intricate neurocircuitry governing appetite and weight regulation connecting the hypothalamus, pituitary gland, brainstem, adipose tissue, pancreas, and gastrointestinal tract. On a background of an increasing prevalence of population-level common obesity, the number of survivors of congenital (eg, septo-optic dysplasia, Prader-Willi syndrome) and acquired (eg, central nervous system tumors) hypothalamic disorders is increasing, thanks to earlier diagnosis and management as well as better oncological therapies. Although to date the discovery of several appetite-regulating peptides has led to the development of a range of targeted molecular therapies for monogenic obesity syndromes, outside of these disorders these discoveries have not translated into the development of efficacious treatments for other forms of HyOb. This review aims to summarize our current understanding of the neuroendocrine physiology of appetite and weight regulation, and explore our current understanding of the pathophysiology of HyOb.
Collapse
Affiliation(s)
- Hoong-Wei Gan
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Manuela Cerbone
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Mehul Tulsidas Dattani
- Department of Endocrinology, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Genetics & Genomic Medicine Research & Teaching Department, University College London Great Ormond Street Institute for Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
4
|
Rovani S, Butler V, Samara-Boustani D, Pinto G, Gonzalez-Briceno L, Nguyen Quoc A, Vermillac G, Stoupa A, Besançon A, Beltrand J, Thalassinos C, Flechtner I, Dassa Y, Viaud M, Arrom-Branas MB, Boddaert N, Puget S, Blauwblomme T, Alapetite C, Bolle S, Doz F, Grill J, Dufour C, Bourdeaut F, Abbou S, Guerrini-Rousseau L, Leruste A, Beccaria K, Polak M, Kariyawasam D. Long-term weight gain in children with craniopharyngioma. Eur J Endocrinol 2024; 190:363-373. [PMID: 38662730 DOI: 10.1093/ejendo/lvae044] [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: 09/28/2023] [Revised: 02/07/2024] [Accepted: 03/13/2024] [Indexed: 05/09/2024]
Abstract
OBJECTIVE Adamantinomatous craniopharyngioma mainly affects children. Excessive weight gain is a major long-term complication. The primary objective of this study was to assess long-term weight changes in children treated for craniopharyngioma. The secondary objectives were to identify risk factors for excessive weight gain and to look for associations with hypothalamic damage by the tumour or treatment. DESIGN Single-centre retrospective cohort study. METHOD Children managed for craniopharyngioma at our centre between 1990 and 2019 were included. The body mass index (BMI) standard deviation scores (SDS) at baseline and at last follow-up were compared. Univariate and multivariate analyses were performed in order to identify variables associated with the long-term BMI-SDS variation. RESULTS The 108 patients had a mean follow-up of 10.4 years. The mean BMI-SDS increase over time was 2.11 (P < .001) overall, 1.21 (P < .001) in the group without hypothalamic involvement by the tumour, and 1.95 (P < .001) in the group managed using intended hypothalamus-sparing surgery. The absence of hypothalamic involvement by the tumour or treatment was significantly associated with less weight gain (P = .046 and P < .01, respectively). After adjustment, factors associated with a BMI-SDS change greater than 2 were female sex (P = .023), tumour involving the hypothalamus (P = .04), and higher baseline BMI (P < .001). CONCLUSION Clinically significant weight gain occurred in nearly all children treated for craniopharyngioma, including those whose hypothalamus was spared by the tumour and intentionally by treatment. However, hypothalamus integrity was associated with less weight gain. Despite hypothalamus-sparing strategies, hypothalamic obesity remains a major concern, indicating a need for novel treatment approaches.
Collapse
Affiliation(s)
- Sibylle Rovani
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Victoria Butler
- Department of Neonatal Medicine of Port Royal, Cochin Hospital, FHU PREMA, AP-HP Centre-Université Paris Cité, Paris 75014, France
| | - Dinane Samara-Boustani
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Graziella Pinto
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Laura Gonzalez-Briceno
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Adrien Nguyen Quoc
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
| | - Gaëlle Vermillac
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Athanasia Stoupa
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Alix Besançon
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Jacques Beltrand
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
- Cochin Institute, INSERM Department U1016, Paris 75014, France
- IMAGINE Institute Affiliate, INSERM Department U1163, Paris 75015, France
| | - Caroline Thalassinos
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Isabelle Flechtner
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Yamina Dassa
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Magali Viaud
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Maria Beatriz Arrom-Branas
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Nathalie Boddaert
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
- Department of Pediatric Radiology, Hospital Necker Enfants Malades, Université Paris Cité, Paris F-75015, France
| | - Stéphanie Puget
- Department of Neurosurgery, Centre Hospitalier Universitaire de Fort de France, University of Antilles, Fort-de-France 97100, Martinique
| | - Thomas Blauwblomme
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
- Department of Pediatric Neurosurgery, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Claire Alapetite
- Radiation Oncology Department, Curie Institute, Paris 75005, France
- Department ICPO (Institut Curie-Centre de Protonthérapie d'Orsay), Orsay 94800, France
| | - Stéphanie Bolle
- Department SIREDO Center (Care, Innovation, Research in, Children, Adolescent and Young Adults Oncology), Curie Institute, 75005 Paris, France
- Department of Radiation Oncology, Gustave Roussy Institute, Villejuif 94800, France
| | - François Doz
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
- Radiation Department, Proton Center, Orsay 94800, France
| | - Jacques Grill
- Child and Adolescent Cancer Department, Gustave Roussy Institute, Villejuif 94800, France
| | - Christelle Dufour
- Child and Adolescent Cancer Department, Gustave Roussy Institute, Villejuif 94800, France
| | | | - Samuel Abbou
- Child and Adolescent Cancer Department, Gustave Roussy Institute, Villejuif 94800, France
| | - Léa Guerrini-Rousseau
- Child and Adolescent Cancer Department, Gustave Roussy Institute, Villejuif 94800, France
| | - Amaury Leruste
- Radiation Department, Proton Center, Orsay 94800, France
| | - Kévin Beccaria
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
- Department of Pediatric Neurosurgery, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
| | - Michel Polak
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
- Cochin Institute, INSERM Department U1016, Paris 75014, France
- IMAGINE Institute Affiliate, INSERM Department U1163, Paris 75015, France
| | - Dulanjalee Kariyawasam
- Paediatric Endocrinology, Diabetology, Gynaecology Department, Necker-Enfants Malades University Hospital, AP-HP Centre, Paris 75015, France
- Faculty of Medicine, Université Paris Cité, Paris 75006, France
- Cochin Institute, INSERM Department U1016, Paris 75014, France
- IMAGINE Institute Affiliate, INSERM Department U1163, Paris 75015, France
| |
Collapse
|
5
|
Roth CL, McCormack SE. Acquired hypothalamic obesity: A clinical overview and update. Diabetes Obes Metab 2024; 26 Suppl 2:34-45. [PMID: 38450938 DOI: 10.1111/dom.15530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/08/2024]
Abstract
Hypothalamic obesity (HO) is a rare and complex disorder that confers substantial morbidity and excess mortality. HO is a unique subtype of obesity characterized by impairment in the key brain pathways that regulate energy intake and expenditure, autonomic nervous system function, and peripheral hormonal signalling. HO often occurs in the context of hypothalamic syndrome, a constellation of symptoms that follow from disruption of hypothalamic functions, for example, temperature regulation, sleep-wake circadian control, and energy balance. Genetic forms of HO, including the monogenic obesity syndromes, often impact central leptin-melanocortin pathways. Acquired forms of HO occur as a result of tumours impacting the hypothalamus, such as craniopharyngioma, surgery or radiation to treat those tumours, or other forms of hypothalamic damage, such as brain injury impacting the region. Risk for severe obesity following hypothalamic injury is increased with larger extent of hypothalamic damage or lesions that contain the medial and posterior hypothalamic nuclei that support melanocortin signalling pathways. Structural damage in these hypothalamic nuclei often leads to hyperphagia, central insulin and leptin resistance, decreased sympathetic activity, low energy expenditure, and increased energy storage in adipose tissue, the collective effect of which is rapid weight gain. Individuals with hyperphagia are perpetually hungry. They do not experience fullness at the end of a meal, nor do they feel satiated after meals, leading them to consume larger and more frequent meals. To date, most efforts to treat HO have been disappointing and met with limited, if any, long-term success. However, new treatments based on the distinct pathophysiology of disturbed energy homeostasis in acquired HO may hold promise for the future.
Collapse
Affiliation(s)
- Christian L Roth
- Centre for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, DC, USA
- Department of Paediatrics, University of Washington, School of Medicine, Seattle, Washington, DC, USA
| | - Shana E McCormack
- Neuroendocrine Centre, Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Paediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Hammoud RA, Mulrooney DA, Rhea IB, Yu C, Johnson JN, Chow EJ, Ehrhardt MJ, Hudson MM, Ness KK, Armstrong GT, Dixon SB. Modifiable Cardiometabolic Risk Factors in Survivors of Childhood Cancer: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:16-32. [PMID: 38510292 PMCID: PMC10950443 DOI: 10.1016/j.jaccao.2023.12.008] [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: 08/03/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 03/22/2024] Open
Abstract
The growing community of childhood cancer survivors faces a heavy burden of late onset morbidities and mortality, with cardiovascular diseases being the leading noncancer cause. In addition to demographics and cancer treatment exposures, which cannot be altered, cardiometabolic risk factors (obesity, hypertension, diabetes, and dyslipidemia) and frailty potentiate the risk of morbidity and mortality associated with chronic health conditions. Important opportunities exist to target these risk factors and improve late health outcomes for survivors. Unfortunately, limited evidence exists on the optimal methods to prevent, screen, and treat cardiometabolic risk factors among survivors, resulting in significant underdiagnosis and undertreatment. In this review, we discuss the prevalence of, risk factors for, current survivor-specific recommendations, and gaps in knowledge to mitigate potentially modifiable cardiometabolic risk factors and frailty among survivors of childhood cancer.
Collapse
Affiliation(s)
- Rawan A. Hammoud
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Daniel A. Mulrooney
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Isaac B. Rhea
- Department of Internal Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christine Yu
- Department of Pediatric Medicine, Division of Endocrinology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason N. Johnson
- Division of Pediatric Cardiology, University of Tennessee Health Science Center, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - Eric J. Chow
- Fred Hutchinson Cancer Center, Seattle Children’s Hospital, University of Washington, Seattle, Washington, USA
| | - Matthew J. Ehrhardt
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Kirsten K. Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gregory T. Armstrong
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stephanie B. Dixon
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
7
|
Hong CS, Smith TR. Aerobic exercise interventions to address impaired quality of life in patients with pituitary tumors. PLoS One 2023; 18:e0295907. [PMID: 38100429 PMCID: PMC10723697 DOI: 10.1371/journal.pone.0295907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
Patients with pituitary tumors may experience persistent fatigue and reduced physical activity, based on subjective measures after treatment. These symptoms may persist despite gross total resection of their tumors and biochemical normalization of pituitary function. While reduced quality of life has been commonly acknowledged in pituitary tumor patients, there is a lack of studies on what interventions may be best implemented to ameliorate these issues, particularly when hormonal levels have otherwise normalized. Aerobic exercise programs have been previously described to ameliorate symptoms of chronic fatigue and reduced physical capacity across a variety of pathologies in the literature. As such, a prescribed aerobic exercise program may be an underrecognized but potentially impactful intervention to address quality of life in pituitary tumor patients. This review seeks to summarize the existing literature on aerobic exercise interventions in patients with pituitary tumors. In addition, future areas of study are discussed, including tailoring exercise programs to the hormonal status of the patient and incorporating more objective measures in monitoring response to interventions.
Collapse
Affiliation(s)
- Christopher S. Hong
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Computational Neuroscience Outcomes Center (CNOC), Boston, Masachusettts, United States of America
| | - Timothy R. Smith
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Computational Neuroscience Outcomes Center (CNOC), Boston, Masachusettts, United States of America
| |
Collapse
|
8
|
Roth CL, Zenno A. Treatment of hypothalamic obesity in people with hypothalamic injury: new drugs are on the horizon. Front Endocrinol (Lausanne) 2023; 14:1256514. [PMID: 37780616 PMCID: PMC10533996 DOI: 10.3389/fendo.2023.1256514] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/22/2023] [Indexed: 10/03/2023] Open
Abstract
Hypothalamic obesity (HO) is a complex and rare disorder affecting multiple regulatory pathways of energy intake and expenditure in the brain as well as the regulation of the autonomic nervous system and peripheral hormonal signaling. It can be related to monogenic obesity syndromes which often affect the central leptin-melanocortin pathways or due to injury of the hypothalamus from pituitary and hypothalamic tumors, such as craniopharyngioma, surgery, trauma, or radiation to the hypothalamus. Traditional treatments of obesity, such as lifestyle intervention and specific diets, are still a therapeutic cornerstone, but often fail to result in meaningful and sustained reduction of body mass index. This review will give an update on pharmacotherapies of HO related to hypothalamic injury. Recent obesity drug developments are promising for successful obesity intervention outcomes.
Collapse
Affiliation(s)
- Christian L. Roth
- Seattle Children’s Research Institute, Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, United States
- Division of Endocrinology, Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Anna Zenno
- Division of Endocrinology, Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
9
|
Shoemaker AH, Tamaroff J. Approach to the Patient With Hypothalamic Obesity. J Clin Endocrinol Metab 2023; 108:1236-1242. [PMID: 36413492 PMCID: PMC10306088 DOI: 10.1210/clinem/dgac678] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Hypothalamic obesity (HO) is defined as abnormal weight gain due to physical destruction of the hypothalamus. Suprasellar tumors, most commonly craniopharyngiomas, are a classic cause of HO. HO often goes unnoticed initially as patients, families, and medical teams are focused on oncologic treatments and management of panhypopituitarism. HO is characterized by rapid weight gain in the first year after hypothalamic destruction followed by refractory obesity due to an energy imbalance of decreased energy expenditure without decreased food intake. Currently available pharmacotherapies are less effective in HO than in common obesity. While not a cure, dietary interventions, pharmacotherapy, and bariatric surgery can mitigate the effects of HO. Early recognition of HO is necessary to give an opportunity to intervene before substantial weight gain occurs. Our goal for this article is to review the pathophysiology of HO and to discuss available treatment options and future directions for prevention and treatment.
Collapse
Affiliation(s)
- Ashley H Shoemaker
- Pediatric Endocrinology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Jaclyn Tamaroff
- Pediatric Endocrinology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
10
|
Huynh K, Klose M, Krogsgaard K, Drejer J, Byberg S, Madsbad S, Magkos F, Aharaz A, Edsberg B, Tfelt-Hansen J, Astrup AV, Feldt-Rasmussen U. Randomized controlled trial of Tesomet for weight loss in hypothalamic obesity. Eur J Endocrinol 2022; 186:687-700. [PMID: 35294397 PMCID: PMC9175551 DOI: 10.1530/eje-21-0972] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/16/2022] [Indexed: 11/21/2022]
Abstract
CONTEXT Hypothalamic injury often leads to rapid, intractable weight gain causing hypothalamic obesity, which is associated with increased risk of cardiovascular and metabolic morbidity and mortality. There are no approved or effective pharmacological treatments for hypothalamic obesity, and conventional lifestyle management remains ineffective. OBJECTIVE To investigate the safety and efficacy of Tesomet (0.5 mg tesofensine/50 mg metoprolol) in adults with hypothalamic obesity. METHODS Twenty-one adults with hypothalamic obesity (16 females) were randomized to Tesomet (0.5 mg/50 mg) or placebo for 24 weeks. Patients also received diet/lifestyle counselling. The primary endpoint was safety; secondary endpoints included measures of body weight, appetite scores, quality of life, and metabolic profile. RESULTS Eighteen patients completed 24 weeks. Consent withdrawal, eligibility, and serious adverse events (SAE) unrelated to treatment resulted in dropouts. One patient experienced a Tesomet-related SAE of exacerbated pre-existing anxiety leading to treatment discontinuation. Tesomet-related adverse events were otherwise mostly mild and included sleep disturbances (Tesomet 50%, placebo 13%), dry mouth (Tesomet 43%, placebo 0%), and headache (Tesomet 36%, placebo 0%). No significant differences in heart rate or blood pressure were observed between groups. Compared to placebo, Tesomet resulted in additional mean (95% CI) weight change of -6.3% ((-11.3; -1.3); P = 0.017), increased the number of patients achieving ≥5% weight loss (Tesomet 8/13, placebo 1/8; P = 0.046), and tended to augment the reduction in waist circumference by 5.7 cm ((-0.1; 11.5); P = 0.054). CONCLUSION Tesomet was welltolerated, did not affect heart rate or blood pressure, and resulted in significant reductions in body weight compared to placebo in adults with hypothalamic obesity.
Collapse
Affiliation(s)
- Kim Huynh
- Department of Medical Endocrinology and Metabolism PE 2131/2, Copenhagen University Hospital (Rigshospitalet), Copenhagen Ø, Denmark
| | - Marianne Klose
- Department of Medical Endocrinology and Metabolism PE 2131/2, Copenhagen University Hospital (Rigshospitalet), Copenhagen Ø, Denmark
| | | | | | - Sarah Byberg
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre University Hospital, University of Copenhagen, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Clinical Sciences, Copenhagen, Copenhagen N, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports (NEXS), University of Copenhagen, Frederiksberg C, Denmark
| | | | | | - Jacob Tfelt-Hansen
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital (Rigshospitalet), Copenhagen Ø, Denmark
- Department of Forensic Medicine, Faculty of Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Vernon Astrup
- Department of Nutrition, Exercise and Sports (NEXS), University of Copenhagen, Frederiksberg C, Denmark
| | - Ulla Feldt-Rasmussen
- Department of Medical Endocrinology and Metabolism PE 2131/2, Copenhagen University Hospital (Rigshospitalet), Copenhagen Ø, Denmark
- Department of Clinical Medicine, Faculty of Health and Clinical Sciences, Copenhagen, Copenhagen N, Denmark
- Correspondence should be addressed to U Feldt-Rasmussen;
| |
Collapse
|
11
|
Dimitri P. Treatment of Acquired Hypothalamic Obesity: Now and the Future. Front Endocrinol (Lausanne) 2022; 13:846880. [PMID: 35464063 PMCID: PMC9019363 DOI: 10.3389/fendo.2022.846880] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
The hypothalamus is the centre of neuroendocrine regulation of energy homeostasis and appetite. Maldevelopment of, or damage to, the key hypothalamic nuclei disrupts the coordinated balance between energy intake and expenditure leading, to rapid and excessive weight gain. Hypothalamic obesity is compounded by a disruption of the hypothalamic-pituitary axis, sleep disruption, visual compromise, and neurological and vascular sequalae. Amongst suprasellar tumors, craniopharyngioma is the most common cause of acquired hypothalamic obesity, either directly or following surgical or radiotherapeutic intervention. At present, therapy is limited to strategies to manage obesity but with a modest and variable impact. Current approaches include optimizing pituitary hormone replacement, calorie restriction, increased energy expenditure through physical activity, behavioral interventions, pharmacotherapy and bariatric surgery. Current pharmacotherapeutic approaches include stimulants that increase energy consumption, anti-diabetic agents, hypothalamic-pituitary substitution therapy, octreotide, and methionine aminopeptidase 2 (MetAP2) inhibitors. Some pharmacological studies of hypothalamic obesity report weight loss or stabilization but reported intervention periods are short, and others report no effect. The impact of bariatric surgery on weight loss in hypothalamic obesity again is variable. Novel or combined approaches to manage hypothalamic obesity are thus required to achieve credible and sustained weight loss. Identifying etiological factors contributing hypothalamic obesity may lead to multi-faceted interventions targeting hyperphagia, insulin resistance, decreased energy expenditure, sleep disturbance, hypopituitarism and psychosocial morbidity. Placebo-controlled trials using current single, or combination therapies are required to determine the impact of therapeutic agents. A well-defined approach to defining the location of hypothalamic damage may support the use of future targeted therapies. Intranasal oxytocin is currently being investigated as an anorexogenic agent. Novel agents including those targeting pro-opimelanocortin-C and AgRP/NPY expressing neurons and the MC4 receptor may result in better outcomes. This article discusses the current challenges in the management of hypothalamic obesity in children and young people and future therapeutic approaches to increasing weight loss and quality of life in these patients.
Collapse
Affiliation(s)
- Paul Dimitri
- The Department of Paediatric Endocrinology, Sheffield Children’s NHS Foundation Trust, Sheffield, United Kingdom
- College of Health, Wellbeing and Life Sciences, Sheffield Hallam University, Sheffield, United Kingdom
- *Correspondence: Paul Dimitri,
| |
Collapse
|
12
|
Perez FA, Elfers C, Yanovski JA, Shoemaker AH, Abuzzahab MJ, Roth CL. MRI measures of hypothalamic injury are associated with glucagon-like peptide-1 receptor agonist treatment response in people with hypothalamic obesity. Diabetes Obes Metab 2021; 23:1532-1541. [PMID: 33651438 PMCID: PMC8353597 DOI: 10.1111/dom.14366] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/15/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
AIM To evaluate whether neuroimaging-delineated regions of hypothalamic injury are associated with a differential treatment response to a glucagon-like peptide-1 receptor agonist (GLP-1RA) in patients with hypothalamic obesity (HO). MATERIALS AND METHODS We performed a prespecified secondary analysis of a randomized, multicentre, double-blind, placebo-controlled trial of people aged 10-25 years with hypothalamic injury and HO randomized to the GLP-1RA exenatide once-weekly (ExQW) or placebo for 36 weeks. Subjects underwent MRI prior to enrolment and the degree of hypothalamic damage was assessed using an integrative hypothalamic lesion score (HLS). Mammillary body (MB) damage was specifically determined. The main clinical endpoints were % change in body mass index (BMI) and change in % body fat. Nested ANCOVA models including a treatment × imaging measure interaction were compared using partial F-tests to assess whether the effect of ExQW treatment differed by severity of hypothalamic damage. RESULTS Complete data were available in 35/42 randomized participants (placebo, n = 15; ExQW, n = 20). ExQW-treated patients with worse HLS or bilateral MB damage had greater reductions in % body fat at 36 weeks (interaction coefficient estimates for HLS: -0.9%, 95% CI -1.6% to -0.2%, p = .02; for MB damage: -7.4%, 95% CI -10.1% to -4.7%, p < .001, respectively) but not for BMI % change. Similarly, patients with more damaged and smaller MB cross-sectional areas had greater reductions in % body fat following ExQW (interaction coefficient estimate 0.3%, 95% CI 0.2%-0.4%, p < .001). CONCLUSIONS In people with HO, greater hypothalamic damage as determined by MRI, in particular MB injury, is associated with greater reductions in adiposity following GLP-1RA treatment.
Collapse
Affiliation(s)
- Francisco A. Perez
- Department of Radiology, Seattle Children’s and University of Washington, Seattle, WA 98105
| | - Clinton Elfers
- Seattle Children’s Research Institute, 1900 Ninth Ave, Seattle, WA 98101
| | - Jack A. Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National, Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Ashley H. Shoemaker
- Vanderbilt University Medical Center, Division of Pediatric Endocrinology, Nashville, TN 37212
| | - M. Jennifer Abuzzahab
- McNeely Pediatric Diabetes Center and Endocrinology Clinic, Children’s Minnesota, St. Paul, MN
| | - Christian L. Roth
- Seattle Children’s Research Institute, 1900 Ninth Ave, Seattle, WA 98101
- Department of Pediatrics, University of Washington, 1959 NE Pacific St, Seattle, WA 98195
| |
Collapse
|
13
|
Takami H, Velásquez C, Asha MJ, Oswari S, Almeida JP, Gentili F. Creative and Innovative Methods and Techniques for the Challenges in the Management of Adult Craniopharyngioma. World Neurosurg 2021; 142:601-610. [PMID: 32987616 DOI: 10.1016/j.wneu.2020.05.173] [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: 04/11/2019] [Accepted: 04/05/2020] [Indexed: 10/23/2022]
Abstract
Craniopharyngioma remains a major challenge in daily clinical practice. The pathobiology of the tumor is still elusive, and there are no consensus or treatment guidelines on the optimal management strategy for this relatively rare tumor. However, recent technical and scientific advances, including genomic and radiomic profiling, innovation in surgical approaches, more precise radiotherapy protocols, targeted therapy, and restoration of lost functions all have the potential to significantly improve the outcome of patients with craniopharyngioma in the near future. Although many of these innovative tools in the new armamentarium of the clinician are still in their infancy, they could reduce craniopharyngioma-related morbidity and mortality and improve the patients' quality of life. In this article, we discuss these creative and innovative approaches that may offer solutions to the obstacles faced in treating craniopharyngioma and future possibilities in improving the care of these patients.
Collapse
Affiliation(s)
- Hirokazu Takami
- Department of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Carlos Velásquez
- Department of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Mohammed J Asha
- Department of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Selfy Oswari
- Department of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Joao Paulo Almeida
- Department of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Fred Gentili
- Department of Neurosurgery, Toronto Western Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
14
|
Abstract
Energy homeostasis, appetite, and satiety are modulated by a complex neuroendocrine system regulated by the hypothalamus. Dysregulation of this system resulting in hypothalamic obesity (HO) is caused by brain tumors, neurosurgery, and/or cranial irradiation. Craniopharyngioma (CP) is a paradigmatic disease with regard to the development of HO. Initial hypothalamic involvement of CP and/or treatment-related damage to hypothalamic-pituitary axes result in HO. Attempts to control HO with lifestyle interventions have not been satisfactory. No generally accepted pharmacologic or bariatric therapy for HO in CP has been effective in randomized controlled trials. Accordingly, prevention of HO is recommended.
Collapse
Affiliation(s)
- Hermann L Müller
- Department of Pediatrics and Pediatric Hematology/Oncology, University Children's Hospital, Klinikum Oldenburg AöR, Rahel-Straus-Strasse 10, Oldenburg 26133, Germany.
| |
Collapse
|
15
|
Rebello CJ, Greenway FL. Obesity medications in development. Expert Opin Investig Drugs 2020; 29:63-71. [PMID: 31847611 PMCID: PMC6990416 DOI: 10.1080/13543784.2020.1705277] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022]
Abstract
Introduction: Obesity is compounded by a neurobiology that is resistant to weight loss. Therefore, the development of pharmacotherapies to address the pathology underlying the dysregulation of energy homeostasis is critical.Areas covered: This review examines selected clinical trial evidence for the pharmacologic treatment of obesity and provides an expert opinion on anti-obesity drug development. The article includes the outcomes of anti-obesity medications that have been evaluated in clinical trials but have not yet received approval from the U.S. Food and Drug Administration. The mechanisms of action of glucagon-like peptide-1 agonists and co-agonists, diabetes medications being investigated for weight loss, and medications acting on the central nervous system as well as peripherally are reviewed. A search was conducted on PubMed using the terms 'Obesity AND Medications' restricted to clinical trials reported in English. Using similar terms, a search was also conducted on ClinicalTrials.gov.Expert opinion: The goal of anti-obesity therapy is finding compounds that are effective and have minimal side effects. Combining medications targeting more than one of the redundant mechanisms driving obesity increases efficacy. However, targeting peripheral mechanisms to overcome the trickle-down effects of centrally acting drugs may be the key to success in treating obesity.
Collapse
Affiliation(s)
- Candida J. Rebello
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Frank L. Greenway
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
16
|
Siddik MAB, Das BC, Weiss L, Dhurandhar NV, Hegde V. A MetAP2 inhibitor blocks adipogenesis, yet improves glucose uptake in cells. Adipocyte 2019; 8:240-253. [PMID: 31264515 PMCID: PMC6768232 DOI: 10.1080/21623945.2019.1636627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue expansion involves angiogenesis to remodel its capillary network. The enzymemethionine aminopeptidase 2(MetAP2) promotes angiogenesis.MetAP2 inhibitors suppress angiogenesis and have potential anti-obesity effect. However, impairment in adipose tissue expansion is also linked with impaired glycemic control.This study investigated the effect of BL6, a MetAP2 inhibitor, on adipogenesis and glucose disposal.To test effect on angiogenesis, Human Umbilical Vein Endothelial Cells(HUVECs) were treated with BL6 for 24h to determine tube formation. Further, to test effect on adipogenesis and glucose disposal,3T3-L1 pre-adipocytes were treated with BL6(0 µM, 20µM, 50 µM or 100µM) during differentiation. Differentiated cells were stained with Oil Red O for determining lipid accumulation, and glucose uptake assay. Protein levels and RNA expression for key genes involved in the adipogenic cascade were determined.BL6 treatment of HUVECs dose dependently blocked angiogenesis. During differentiation of pre-adipocytes, 50μM and 100µM BL6 significantly reduced lipid accumulation. Treatment with 100µM BL6 significantly decreased expression of adipogenic genes. Interestingly, BL6 treatment dose dependently increased glucose uptake by 3T3-L1 cells.MetAP2 inhibitor blocks angiogenesis, attenuates adipogenesis, yet increases cellular glucose uptake. Collectively this proof of concept study supports a possible role for MetAP2 inhibitor BL6, as a putative anti-obesity therapeutic agent.
Collapse
Affiliation(s)
| | - Bhaskar C. Das
- The Icahn School of Medicine, Department of Medicine, New York, NY, USA
| | - Louis Weiss
- Department of Pathology, The Albert Einstein College of Medicine, New York, NY, USA
| | | | - Vijay Hegde
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
17
|
Huang HJ, Holub C, Rolzin P, Bilakovics J, Fanjul A, Satomi Y, Plonowski A, Larson CJ, Farrell PJ. MetAP2 inhibition increases energy expenditure through direct action on brown adipocytes. J Biol Chem 2019; 294:9567-9575. [PMID: 31048375 DOI: 10.1074/jbc.ra118.007302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 04/26/2019] [Indexed: 11/06/2022] Open
Abstract
Inhibitors of methionine aminopeptidase 2 (MetAP2) have been shown to reduce body weight in obese mice and humans. The target tissue and cellular mechanism of MetAP2 inhibitors, however, have not been extensively examined. Using compounds with diverse chemical scaffolds, we showed that MetAP2 inhibition decreases body weight and fat mass and increases lean mass in the obese mice but not in the lean mice. Obesity is associated with catecholamine resistance and blunted β-adrenergic receptor signaling activities, which could dampen lipolysis and energy expenditure resulting in weight gain. In the current study, we examined effect of MetAP2 inhibition on brown adipose tissue and brown adipocytes. Norepinephrine increases energy expenditure in brown adipose tissue by providing fatty acid substrate through lipolysis and by increasing expression of uncoupled protein-1 (UCP1). Metabolomic analysis shows that in response to MetAP2 inhibitor treatment, fatty acid metabolites in brown adipose tissue increase transiently and subsequently decrease to basal or below basal levels, suggesting an effect on fatty acid metabolism in this tissue. Treatment of brown adipocytes with MetAP2 inhibitors enhances norepinephrine-induced lipolysis and energy expenditure, and prolongs the activity of norepinephrine to increase ucp1 gene expression and energy expenditure in norepinephrine-desensitized brown adipocytes. In summary, we showed that the anti-obesity activity of MetAP2 inhibitors can be mediated, at least in part, through direct action on brown adipocytes by enhancing β-adrenergic-signaling-stimulated activities.
Collapse
Affiliation(s)
| | - Corine Holub
- From Takeda California, San Diego, California 92121
| | - Paul Rolzin
- From Takeda California, San Diego, California 92121
| | | | | | - Yoshinori Satomi
- Takeda Pharmaceutical Company Limited, Fujisawa 251-0012 Japan, and
| | | | - Christopher J Larson
- From Takeda California, San Diego, California 92121.,Sanford Burham Prebys Medical Discovery Institute, San Diego, California 92037
| | | |
Collapse
|
18
|
Thompson CJ, Costello RW, Crowley RK. Management of hypothalamic disease in patients with craniopharyngioma. Clin Endocrinol (Oxf) 2019; 90:506-516. [PMID: 30614015 DOI: 10.1111/cen.13929] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 12/21/2022]
Abstract
Patients with craniopharyngioma experience excess morbidity and mortality when compared with the background population and with other hypopituitary patients. Large, suprasellar tumours which form micropapillae into surrounding structures can cause hypothalamic damage before any therapeutic intervention; attempted gross total resection can lead to hypothalamic obesity, sleep disorders, thirst disorders and dysregulation of temperature as well as panhypopituitarism. The management of tumour bulk and the pathophysiology of hypothalamic complications have been reviewed extensively. We present a practical, clinical approach to management of hypothalamic disease in a patient with craniopharyngioma and highlight potential targets for future pharmacological or surgical intervention.
Collapse
Affiliation(s)
- Christopher J Thompson
- Academic Department of Endocrinology, Beaumont Hospital, Dublin, Ireland
- Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Richard W Costello
- Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
- Department of Respiratory Medicine, Beaumont Hospital, Dublin, Ireland
| | - Rachel K Crowley
- Department of Endocrinology, St Vincent's University Hospital, Dublin, Ireland
- University College, Dublin, Ireland
| |
Collapse
|
19
|
van Iersel L, Brokke KE, Adan RAH, Bulthuis LCM, van den Akker ELT, van Santen HM. Pathophysiology and Individualized Treatment of Hypothalamic Obesity Following Craniopharyngioma and Other Suprasellar Tumors: A Systematic Review. Endocr Rev 2019; 40:193-235. [PMID: 30247642 DOI: 10.1210/er.2018-00017] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 06/25/2018] [Indexed: 12/26/2022]
Abstract
The development of hypothalamic obesity (HO) following craniopharyngioma (CP) and other suprasellar tumors leads to reduced patient quality of life. No treatment algorithms are currently available for management of HO. Depending on which hypothalamic nuclei are destroyed, the pathophysiologic mechanisms and clinical symptoms that contribute to HO differ among patients. Herein, we review the contribution of the hypothalamus to the pathophysiologic mechanisms and symptoms underlying CP-associated HO. Additionally, we performed a systematic search of MEDLINE and Embase to identify all intervention studies for weight management in patients with CP or other suprasellar tumors published until September 2017. The search yielded 1866 publications, of which 40 were included. Of these 40 studies, we identified four modalities for intervention (i.e., lifestyle, dietary, pharmacotherapeutic, or surgical) within six clinical domains (i.e., psychosocial disorders, hyperphagia, sleep disturbances, decreased energy expenditure, hyperinsulinemia, and hypopituitarism). We used the findings from our systematic review, in addition to current knowledge on the pathophysiology of HO, to develop an evidence-based treatment algorithm for patients with HO caused by CP or other suprasellar tumors. Although the individual effects of the HO interventions were modest, beneficial individual effects may be achieved when the pathophysiologic background and correct clinical domain are considered. These two aspects can be combined in an individualized treatment algorithm with a stepwise approach for each clinical domain. Recently elucidated targets for HO intervention were also explored to improve future management of HO for patients with CP and other suprasellar tumors.
Collapse
Affiliation(s)
- Laura van Iersel
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Karen E Brokke
- Medical Sciences, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Roger A H Adan
- Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, Netherlands.,Institute for Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lauren C M Bulthuis
- Medical Sciences, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Erica L T van den Akker
- Department of Pediatric Endocrinology, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Hanneke M van Santen
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
20
|
Bogusz A, Müller HL. Childhood-onset craniopharyngioma: latest insights into pathology, diagnostics, treatment, and follow-up. Expert Rev Neurother 2018; 18:793-806. [DOI: 10.1080/14737175.2018.1528874] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Agnieszka Bogusz
- Department of Pediatrics and Pediatric Hematology/Oncology, Klinikum Oldenburg AöR, Medical Campus University Oldenburg, Oldenburg, Germany
- Department of Endocrinology and Diabetology, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Hermann L. Müller
- Department of Pediatrics and Pediatric Hematology/Oncology, Klinikum Oldenburg AöR, Medical Campus University Oldenburg, Oldenburg, Germany
| |
Collapse
|
21
|
Proietto J, Malloy J, Zhuang D, Arya M, Cohen ND, de Looze FJ, Gilfillan C, Griffin P, Hall S, Nathow T, Oldfield GS, O'Neal DN, Roberts A, Stuckey BGA, Yue D, Taylor K, Kim D. Efficacy and safety of methionine aminopeptidase 2 inhibition in type 2 diabetes: a randomised, placebo-controlled clinical trial. Diabetologia 2018; 61:1918-1922. [PMID: 29992370 DOI: 10.1007/s00125-018-4677-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/24/2018] [Indexed: 10/28/2022]
Abstract
AIMS/HYPOTHESIS This multicentre randomised double-blind placebo-controlled clinical trial assessed the efficacy and safety of a methionine aminopeptidase 2 (MetAP2) inhibitor, beloranib, in individuals with obesity (BMI ≥30 kg/m2) and type 2 diabetes (HbA1c 53-97 mmol/mol [7-11%] and fasting glucose <15.6 mmol/l). METHODS Participants were randomised (via a centralised interactive web response system) to placebo, 1.2 or 1.8 mg beloranib s.c. twice weekly for 26 weeks. Participants, investigators and the sponsor were blinded to group assignment. The primary endpoint was the change in weight from baseline to week 26. The trial was terminated early when beloranib development was stopped because of an imbalance of venous thromboembolism events in beloranib-treated individuals vs placebo that became evident during late-stage development of the drug. RESULTS In total, 153 participants were randomised, 51 to placebo, 52 to 1.2 mg beloranib and 50 to 1.8 mg beloranib. In participants who completed week 26, the least squares mean ± SE weight change (baseline 111 kg) was -3.1 ± 1.2% with placebo (n = 22) vs -13.5 ± 1.1% and -12.7 ± 1.3% with 1.2 and 1.8 mg beloranib, respectively (n = 25; n = 19; p < 0.0001). The change in HbA1c (baseline 67 mmol/mol [8.3%]) was -6.6 ± 2.2 mmol/mol (-0.6 ± 0.2%) with placebo vs -21.9 ± 2.2 mmol/mol (-2.0 ± 0.2%) or -21.9 ± 3.3 mmol/mol (-2.0 ± 0.3%) with 1.2 or 1.8 mg beloranib (p < 0.0001), respectively. The most common beloranib adverse events were sleep related. One beloranib-treated participant experienced a non-fatal pulmonary embolism. CONCLUSIONS/INTERPRETATION MetAP2 inhibitors represent a novel mechanism for producing meaningful weight loss and improvement in HbA1c. TRIAL REGISTRATION ClinicalTrials.gov NCT02324491 FUNDING: The study was funded by Zafgen, Inc.
Collapse
Affiliation(s)
- Joseph Proietto
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, VIC, Australia
| | - Jaret Malloy
- Zafgen, Inc., 175 Portland St, 4th Floor, Boston, MA, 02114, USA
| | - Dongliang Zhuang
- Zafgen, Inc., 175 Portland St, 4th Floor, Boston, MA, 02114, USA
| | - Mark Arya
- Australian Clinical Research Network, Maroubra, NSW, Australia
| | - Neale D Cohen
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ferdinandus J de Looze
- AusTrials Pty Ltd, Sherwood, QLD, Australia
- Discipline of General Practice, Faculty of Medicine, University of Queensland, Herston, QLD, Australia
| | | | - Paul Griffin
- Q-Pharm Pty Ltd, Herston, QLD, Australia
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- Mater Health Services, South Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Herston, QLD, Australia
| | - Stephen Hall
- Emeritus Research, Malvern East, VIC, Australia
- Institution for Rehabilitation Research, Monash University, Clayton, VIC, Australia
| | | | | | - David N O'Neal
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Adam Roberts
- University Hospital Geelong, Geelong, VIC, Australia
| | - Bronwyn G A Stuckey
- Keogh Institute for Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - Dennis Yue
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Kristin Taylor
- Zafgen, Inc., 175 Portland St, 4th Floor, Boston, MA, 02114, USA
| | - Dennis Kim
- Zafgen, Inc., 175 Portland St, 4th Floor, Boston, MA, 02114, USA.
| |
Collapse
|
22
|
Malloy J, Zhuang D, Kim T, Inskeep P, Kim D, Taylor K. Single and multiple dose evaluation of a novel MetAP2 inhibitor: Results of a randomized, double-blind, placebo-controlled clinical trial. Diabetes Obes Metab 2018; 20:1878-1884. [PMID: 29577550 PMCID: PMC6055687 DOI: 10.1111/dom.13305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/11/2018] [Accepted: 03/20/2018] [Indexed: 11/29/2022]
Abstract
AIMS Methionine aminopeptidase 2 (MetAP2) inhibition has been shown to result in significant weight loss and improved glucose control. This Phase 1 clinical trial assessed the safety and tolerability, pharmacokinetics and preliminary efficacy of a novel MetAP2 inhibitor, ZGN-1061. METHODS This clinical trial included a single ascending dose (SAD) phase in healthy subjects (BMI, 23 to <30 kg/m2 ) and a multiple ascending dose (MAD) phase in otherwise healthy subjects (BMI, 27 to 40 kg/m2 ). SAD phase doses, administered subcutaneously (SC), were 0.2, 0.6, 1.2, 2.4, 3.6 and 4.8 mg and the MAD phase evaluated doses of 0.2, 0.6 and 1.8 mg twice weekly SC for 4 weeks. RESULTS The SAD phase included 39 subjects (ZGN-1061, N = 28; placebo, N = 11); 90% were male and BMI was 26.4 kg/m2 . ZGN-1061 was well tolerated across all doses, with the most frequent adverse events being mild headache and procedural-related irritation. There were no severe or serious adverse events. All doses of ZGN-1061 were rapidly absorbed and cleared, resulting in short duration of exposure that is anticipated to minimize potential off-drug target risks. The MAD phase included 29 subjects (ZGN-1061, N = 22; placebo, N = 7); 76% were male and BMI was 33.5 kg/m2 . Safety observations were consistent with SAD findings. Efficacy measures in the MAD phase indicated trends for weight change (-1.5 kg total ZGN-1061 vs -0.2 kg placebo) and other biomarker changes. CONCLUSIONS ZGN-1061 was well tolerated with no safety signals in all doses tested. In addition, the desired pharmacokinetic profile and preliminary efficacy observations with ZGN-1061 support evaluation in larger and longer clinical trials.
Collapse
|
23
|
Ni W, Shi X. Interventions for the Treatment of Craniopharyngioma-Related Hypothalamic Obesity: A Systematic Review. World Neurosurg 2018; 118:e59-e71. [PMID: 29945001 DOI: 10.1016/j.wneu.2018.06.121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Craniopharyngiomas (CPs) and their treatment are associated with hypothalamic damage that causes hypothalamic obesity (HO) in 30%-70% of cases. Thus, there is ongoing research regarding tangible solutions for HO, because these patients have unrelenting resistance to basic weight-loss interventions. This review aims to summarize the interventions that are used to treat CP-related HO (CP-HO), including pharmacotherapy and bariatric surgery. METHODS The Cochrane Library, EMBASE, and PubMed databases were searched up to June 2017 for relevant reports. Two reviewers conducted independent evaluations of the studies identified. RESULTS Eighteen articles were included in the systematic review, with 3 reports describing pharmacotherapy in randomized controlled trials and 15 reports describing bariatric surgery. Although several studies described effective interventions for treating CP-HO, the evidence base was limited by its low quality and our inability to perform a meta-analysis, which was related to a lack of adequate or integrated data. CONCLUSIONS Octreotide appears to be a preferred treatment for patients with CP-HO, based on limited data. Gastric bypass surgery may also be suitable for select patients with CP-HO, based on a review of various procedures in this setting. Microsurgical preservation of the hypothalamic structures is mandatory to decrease CP-HO-related morbidity and mortality. Further studies with adequate analytical power and sufficient follow-up are needed to identify effective strategies for CP-HO treatment.
Collapse
Affiliation(s)
- Weimin Ni
- Department of Neurosurgery, Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Xiang'en Shi
- Department of Neurosurgery, Fu Xing Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
24
|
Greydanus DE, Agana M, Kamboj MK, Shebrain S, Soares N, Eke R, Patel DR. Pediatric obesity: Current concepts. Dis Mon 2018; 64:98-156. [DOI: 10.1016/j.disamonth.2017.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Burkey BF, Hoglen NC, Inskeep P, Wyman M, Hughes TE, Vath JE. Preclinical Efficacy and Safety of the Novel Antidiabetic, Antiobesity MetAP2 Inhibitor ZGN-1061. J Pharmacol Exp Ther 2018; 365:301-313. [PMID: 29491038 DOI: 10.1124/jpet.117.246272] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/23/2018] [Indexed: 01/12/2023] Open
Abstract
Methionine aminopeptidase 2 (MetAP2) inhibition is a promising approach to treating diabetes, obesity, and associated metabolic disorders. Beloranib, a MetAP2 inhibitor previously investigated for treatment of Prader-Willi syndrome, was associated with venous thrombotic adverse events likely resulting from drug effects on vascular endothelial cells (ECs). Here, we report the pharmacological characterization of ZGN-1061, a novel MetAP2 inhibitor being investigated for treatment of diabetes and obesity. Four weeks of subcutaneous administration of ZGN-1061 to diet-induced obese (DIO) insulin-resistant mice produced a 25% reduction in body weight, primarily due to reduced fat mass, that was comparable to beloranib. ZGN-1061 also produced improvements in metabolic parameters, including plasma glucose and insulin, and, in HepG2 cells, initiated gene changes similar to beloranib that support observed in vivo pharmacodynamics. In vitro studies in ECs demonstrated that ZGN-1061 effects on EC proliferation and coagulation proteins were greatly attenuated, or absent, relative to beloranib, due to lower intracellular drug concentrations, shorter half-life of inhibitor-bound MetAP2 complex, and reduced cellular enzyme inhibition. In dogs, ZGN-1061 was more rapidly absorbed and cleared, with a shorter half-life than beloranib. Unlike beloranib, ZGN-1061 did not increase coagulation markers in dogs, and ZGN-1061 had a greatly improved safety profile in rats relative to beloranib. In conclusion, ZGN-1061 and beloranib demonstrated similar efficacy in a mouse model of obesity, while ZGN-1061 had a markedly improved safety profile in multiple in vitro and in vivo models. The lower duration of exposure characteristic of ZGN-1061 is expected to provide a meaningfully enhanced clinical safety profile.
Collapse
Affiliation(s)
- Bryan F Burkey
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Niel C Hoglen
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Philip Inskeep
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Margaret Wyman
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Thomas E Hughes
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - James E Vath
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| |
Collapse
|