1
|
Castiñeiras Pardines A, López Ginés G, García Orueta G, F Trocóniz I. Development and evaluation of a model characterizing the release characteristics of a new letrozole long-acting injectable formulation. Eur J Pharm Sci 2025; 209:107103. [PMID: 40252852 DOI: 10.1016/j.ejps.2025.107103] [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: 12/12/2024] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Treating a chronic condition such as breast cancer usually requires daily oral drug administration for extended periods of time, which is associated with non-adherence to the prescribed therapy that may cause disease progression. New delivery strategies such as long-acting injectable (LAI) implants have entered the picture in order to solve oral administration drawbacks while improving bioavailability, plasma levels variability or treatment compliance. This has motivated the development of a new polymeric and biodegradable in situ forming long-acting implant of letrozole. This new formulation is provided as a kit of two syringes (one of them containing letrozole and Poly-Lactic Acid, and the other one containing dimethyl sulfoxide as solvent for reconstitution). Once the formulation is reconstituted and injected into the muscle, the solvent diffuses into tissue fluids and the insoluble polymer precipitates, forming a semi-solid implant that traps the API and allows a sustained drug release. In order to optimize both the formulation and the development process, traditional in vitro dissolution assessment and predictive dissolution modelling were conducted to identify which formulation characteristics show an impact on the kinetics of the release, which may provide a first basis to potentially establish an in vitro-in vivo correlation (IVIVC) with both pre-clinical and clinical data in the future. Two dissolution methods (real-time and accelerated) were used to describe the in vitro dissolution profiles of 15 letrozole LAI formulations differing on their Critical Material Attributes (CMAs). The release profiles were best described using the Weibull distribution and estimating the fraction of the dose loss during injection. The first order rate constant of release (KD) was increased by 1.87 times in the case of the accelerated conditions, and was 30 % reduced and increased by 1.34 times in the case of higher and lower viscosity of the formulations, respectively. This work allowed for quantitative characterization of the formulation related characteristics responsible for controlling drug release. It provides a new understanding of the formulation that will serve to guide in the development of a robust formulation and to establish product quality control specifications.
Collapse
Affiliation(s)
- Adriana Castiñeiras Pardines
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; Laboratorios Farmacéuticos Rovi, S.A., Granada, Spain.
| | | | - Gastón García Orueta
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
| | - Iñaki F Trocóniz
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain; IdisNA, Navarra Institute for Health Research, Pamplona, Spain; Institute of Data Science and Artificial Intelligence, DATAI, University of Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Zeger VR, Thapa B, Shamsaei D, DeLair JF, Taylor TL, Anderson JL. Ionic Liquids in Analytical Chemistry: Fundamentals, Technological Advances, and Future Outlook. Anal Chem 2025; 97:4793-4818. [PMID: 40018979 PMCID: PMC11912132 DOI: 10.1021/acs.analchem.5c00264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
|
3
|
Wanselius M, Abrahmsén-Alami S, Hanafy BI, Mazza M, Hansson P. A microfluidic in vitro method predicting the fate of peptide drugs after subcutaneous administration. Int J Pharm 2024; 667:124849. [PMID: 39454976 DOI: 10.1016/j.ijpharm.2024.124849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
For many biopharmaceuticals, subcutaneous (sc) administration is the only viable route. However, there is no in vitro method available accurately predicting the absorption profiles of subcutaneously injected pharmaceuticals. In this work, we show that a recently developed microfluidics method for interaction studies (MIS) has the potential to be useful in this respect. The method utilises the responsiveness of polyelectrolyte microgel networks to oppositely charged molecules as a means to monitor the interaction between peptides and hyaluronic acid (HA), a major constituent of the subcutaneous extracellular matrix. We use the method to determine parameters describing the strength of interaction between peptide and HA as well as the peptide's aggregation tendency and transport properties in HA networks. The results from MIS studies of the peptide drugs exenatide, pramlintide, vancomycin, polymyxin B, lanreotide, MEDI7219 and AZD2820 are compared with results from measurements with the commercially available SCISSOR system and in vivo absorption and bioavailability data from the literature. We show that both MIS and SCISSOR reveal differences in the peptides' diffusivity and tendency to aggregate in the presence of HA. We show that MIS is particularly good at discriminating between peptides forming aggregates stabilised by non-electrostatic forces in the presence of HA, and peptides forming complexes stabilised by electrostatic interactions with HA. The method provides two parameters that can be used to quantify the peptides' aggregation tendency, the one describing the peptide packing density in complexes with HA and the other the apparent diffusivity upon release in a medium of physiological ionic strength and pH. The order of the peptides when ranked by increasing binding strength at pH 7.4 determined with MIS is shown to be in agreement with the order when ranked by the apparent 1st order absorption rate constant (ka) after sc administration in humans: lanreotide (Autogel) < exenatide (IRF) < AZD2820 < pramlintide < lanreotide (IRF) (IRF: Immediate release formulation). A correlation is found between the 1st order release rate constant determined with SCISSOR and ka for lanreotide (Autogel), exenatide and AZD2820. A mechanism relating the magnitude of ka to the peptides' charge is proposed.
Collapse
Affiliation(s)
- Marcus Wanselius
- Department of Medicinal Chemistry, Uppsala University, BMC P.O. Box 574, SE-751 23, Uppsala, Sweden
| | - Susanna Abrahmsén-Alami
- Innovation Strategy & External Liaison. Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Belal I Hanafy
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge, United Kingdom
| | - Mariarosa Mazza
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge, United Kingdom
| | - Per Hansson
- Department of Medicinal Chemistry, Uppsala University, BMC P.O. Box 574, SE-751 23, Uppsala, Sweden.
| |
Collapse
|
4
|
Ruggeri RM, Aini I, Gay S, Grossrubatscher EM, Mancini C, Tarsitano MG, Zamponi V, Isidori AM, Colao A, Faggiano A. Efficacy and tolerability of somatostatin analogues according to gender in patients with neuroendocrine tumors. Rev Endocr Metab Disord 2024; 25:383-398. [PMID: 38051470 DOI: 10.1007/s11154-023-09858-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2023] [Indexed: 12/07/2023]
Abstract
As the incidence of neuroendocrine tumors has been rising, gender differences in epidemiology and clinical behavior have emerged, and interest into a gender-driven management of these tumors has grown with the aim to improve survival and quality of life of these patients. Somatostatin Analogues represent the first line of systemic treatment of both functional and non-functional neuroendocrine tumors, through the expression of somatostatin receptors (SSTRs) in the tumor cells, and proved effective in controlling hormonal hypersecretion and inhibiting tumor growth, improving progression-free survival and overall survival of these patients. Aim of the present review is to investigate any differences by gender in efficacy and safety of SSTS-targeted therapies, that represent the mainstay treatment of neuroendocrine tumors, as they emerge from studies of varying design and intent. Although preclinical studies have provided evidence in favor of differences by gender in tumor expression of SSTR, as well as of the role of sex hormones and related receptors in modulating SSTRs expression and function, the clinical studies conducted so far have not shown substantial differences between males and females in either efficacy or toxicity of SSTR-targeted therapies, even if with sometimes inconsistent results. Moreover, in most studies gender was not a predictor of response to treatment. Studies specifically designed to address this issue are needed to develop gender-specific therapeutic algorithms, improving patients' prognosis and quality of life.
Collapse
Affiliation(s)
- Rosaria M Ruggeri
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV, University of Messina, 98125, Messina, Italy.
| | - Irene Aini
- Endocrinology Unit, Azienda Ospedaliera Universitaria Sassari, Sassari, Italy
| | - Stefano Gay
- Endocrinology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Camilla Mancini
- Unit of Andrology and Endocrinology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00189, Rome, Italy
| | | | - Virginia Zamponi
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Andrea M Isidori
- Policlinico Umberto I, Università Sapienza, Gruppo NETTARE, Rome, Italy
| | - Annamaria Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
- UNESCO Chair "Education for Health and Sustainable Development", Federico II University, Naples, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
5
|
Ntorkou M, Kabir A, Furton KG, Tzanavaras PD, Zacharis CK. Sol-gel Carbowax 20M-zwitterionic ionic liquid composite sorbent-based capsule phase microextraction device combined with HPLC/post-column derivatization for the determination of lanreotide, a human somatostatin analogue in urine. J Chromatogr A 2024; 1717:464674. [PMID: 38290172 DOI: 10.1016/j.chroma.2024.464674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
In this research, a sol-gel Carbowax 20M-zwitterionic ionic liquid composite sorbent-based capsule phase microextraction (CPME) device was developed in combination with liquid chromatography-post column derivatization for the first ever reported determination of a somatostatin analogue - lanreotide in human urine. The sol-gel Carbowax 20M-zwitterionic ionic liquid composite sorbent was encapsulated in the lumen of a polypropylene capillary tube and characterized by FT-IR spectroscopy and SEM with energy dispersive X-ray spectroscopy (EDS). The main steps of the CPME workflow were optimized to obtain high extraction efficiency for the target analyte. After the separation of the analyte on a C8 stationary phase, the peptide was derivatized online with o-phthalaldehyde before the fluorescence detection. The main experimental parameters of CPME and the post-column procedures were systematically investigated and optimized. The method was validated in terms of selectivity, linearity, accuracy, precision, limits of detection (LOD), and limits of quantification (LOQ). The relative bias ranged between 88.8 and 115.6 % for the peptide, while the RSD values for repeatability and intermediate precision were less than 14.3 %. The achieved limit of detection (LOD) was 0.2 μΜ while the limit of quantitation (LOQ) was established as 0.9 μΜ. Finally, the sol-gel Carbowax 20M-zwitterionic ionic liquid composite sorbent-based microextraction capsules were found to be reusable for at least 20 extractions. The developed method presented adequate overall performance, and it could be applied in the analysis of selected peptide in human urine samples.
Collapse
Affiliation(s)
- Marianna Ntorkou
- Laboratory of Pharmaceutical Analysis, School of Pharmacy, Aristotle University of Thessaloniki, GR, Thessaloniki 54124, Greece
| | - Abuzar Kabir
- Department of Chemistry and Biochemistry, International Forensic Research Institute, Florida International University, Miami, FL, USA
| | - Kenneth G Furton
- Department of Chemistry and Biochemistry, International Forensic Research Institute, Florida International University, Miami, FL, USA
| | - Paraskevas D Tzanavaras
- Laboratory of Analytical Chemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, GR, 54124, Greece
| | - Constantinos K Zacharis
- Laboratory of Pharmaceutical Analysis, School of Pharmacy, Aristotle University of Thessaloniki, GR, Thessaloniki 54124, Greece.
| |
Collapse
|
6
|
Pharmacophore-Model-Based Drug Repurposing for the Identification of the Potential Inhibitors Targeting the Allosteric Site in Dengue Virus NS5 RNA-Dependent RNA Polymerase. Viruses 2022; 14:v14081827. [PMID: 36016449 PMCID: PMC9412353 DOI: 10.3390/v14081827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Dengue virus (DENV) is the causative agent of DENV infection. To tackle DENV infection, the development of therapeutic molecules as direct-acting antivirals (DAAs) has been demonstrated as a truly effective approach. Among various DENV drug targets, non-structural protein 5 (NS5)-a highly conserved protein among the family Flaviviridae-carries the RNA-dependent RNA polymerase (DENVRdRp) domain at the C-terminal, and its "N-pocket" allosteric site is widely considered for anti-DENV drug development. Therefore, in this study, we developed a pharmacophore model by utilising 41 known inhibitors of the DENVRdRp domain, and performed model screening against the FDA's approved drug database for drug repurposing against DENVRdRp. Herein, drugs complying with the pharmacophore hypothesis were further processed through standard-precision (SP) and extra-precision (XP) docking scores (DSs) and binding pose refinement based on MM/GBSA binding energy (BE) calculations. This resulted in the identification of four potential potent drugs: (i) desmopressin (DS: -10.52, BE: -69.77 kcal/mol), (ii) rutin (DS: -13.43, BE: -67.06 kcal/mol), (iii) lypressin (DS: -9.84, BE: -67.65 kcal/mol), and (iv) lanreotide (DS: -8.72, BE: -64.7 kcal/mol). The selected drugs exhibited relevant interactions with the allosteric N-pocket of DENVRdRp, including priming-loop and entry-point residues (i.e., R729, R737, K800, and E802). Furthermore, 100 ns explicit-solvent molecular dynamics simulations and end-point binding free energy assessments support the considerable stability and free energy of the selected drugs in the targeted allosteric pocket of DENVRdRp. Hence, these four drugs, repurposed as potent inhibitors of the allosteric site of DENVRdRp, are recommended for further validation using experimental assays.
Collapse
|
7
|
Madan A, Markison S, Betz SF, Krasner A, Luo R, Jochelson T, Lickliter J, Struthers RS. Paltusotine, a novel oral once-daily nonpeptide SST2 receptor agonist, suppresses GH and IGF-1 in healthy volunteers. Pituitary 2022; 25:328-339. [PMID: 35000098 PMCID: PMC8894159 DOI: 10.1007/s11102-021-01201-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 01/28/2023]
Abstract
PURPOSE Evaluate the pharmacodynamics, pharmacokinetics, and safety of paltusotine, an orally bioavailable, nonpeptide, somatostatin receptor subtype 2 (SST2) agonist being developed for the treatment of acromegaly and neuroendocrine tumors. METHODS A randomized, double-blind, placebo-controlled, single center, single and multiple ascending dose phase 1 study was conducted in healthy male volunteers who received (i) single-dose of oral paltusotine 1.25, 2.5, 5, 10, and 20 mg (solution); and 40 and 60 mg (capsules) or (ii) multiple-dose oral paltusotine capsules once daily 5 mg (× 7 days), 10, 20, and 30 mg (× 10 days). Main outcome measures were pharmacodynamics (changes in growth hormone-releasing hormone [GHRH] stimulated growth hormone [GH] and insulin-like growth factor 1 [IGF-1]), pharmacokinetics, safety, and tolerability. RESULTS Single-dose cohorts: n = 41 active, n = 14 placebo. Multiple-dose cohorts: n = 24 active, n = 12 placebo. Paltusotine was well tolerated, orally bioavailable, associated with increased plasma concentrations to doses up to 40 mg, and was eliminated with a half-life of approximately 30 h. Single-dose paltusotine 1.25 to 20 mg suppressed GHRH-stimulated GH secretion by 44% to 93% compared to 15% with placebo. Multiple-dose paltusotine 5 to 30 mg administered once daily for 10 days suppressed IGF-1 by 19% to 37% compared to an increase of 2.4% with placebo. CONCLUSIONS Paltusotine suppresses GH and IGF-1 in a dose-dependent fashion, with a safety profile similar to currently approved SST2 receptor ligands. Paltusotine is a promising once-daily oral nonpeptide SST2 agonist candidate for managing acromegaly and neuroendocrine tumors. TRIAL REGISTRATION NCT03276858, registered September 8, 2017, retrospectively registered.
Collapse
Affiliation(s)
- Ajay Madan
- Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Road, Building 2, San Diego, CA, 92121, USA.
- Crinetics Medical Affairs, Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Rd. Bldg.2, San Diego, CA, 92121, USA.
| | - Stacy Markison
- Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Road, Building 2, San Diego, CA, 92121, USA
| | - Stephen F Betz
- Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Road, Building 2, San Diego, CA, 92121, USA
| | - Alan Krasner
- Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Road, Building 2, San Diego, CA, 92121, USA
| | - Rosa Luo
- Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Road, Building 2, San Diego, CA, 92121, USA
| | - Theresa Jochelson
- Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Road, Building 2, San Diego, CA, 92121, USA
| | | | - R Scott Struthers
- Crinetics Pharmaceuticals, Inc, 10222 Barnes Canyon Road, Building 2, San Diego, CA, 92121, USA
| |
Collapse
|
8
|
Neggers S, Badiu C, Biagetti B, Durand-Gasselin L, Petit A, Petrossians P, Regnault B, Rich D, Shafigullina Z, Shustov S, Vydrych A. Pharmacological and safety profile of a prolonged-release lanreotide formulation in acromegaly. Expert Rev Clin Pharmacol 2021; 14:1551-1560. [PMID: 34664531 DOI: 10.1080/17512433.2021.1986004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Patients with acromegaly require lifelong medication; a longer dosing interval would reduce treatment burden. This study investigated the pharmacokinetics, pharmacodynamics and safety profile of a new prolonged-release formulation (PRF) of lanreotide every 12 weeks. RESEARCH DESIGN AND METHODS In this multicenter, open-label, dose-ascending study, cohorts of nine patients with acromegaly received single doses of lanreotide PRF according to a 3 + 3 + 3 scheme in order to determine the maximum tolerated dose (MTD). Following a 12-week treatment period, patients were followed up for a further 12 weeks. Serum lanreotide, insulin-like growth factor-1 and growth hormone concentrations were analyzed. Adverse events were monitored throughout the study. RESULTS The MTD was not reached. Peak lanreotide serum concentration values were similar in all cohorts, whereas area under the curve values from time zero to 85 days increased but were not dose-proportional. The apparent elimination half-life of lanreotide PRF was approximately 54-63 days, in line with the expected prolonged-release characteristics. Growth hormone and insulin-like growth factor-1 levels were generally stable. CONCLUSIONS The safety and tolerability profile was in-line with the known safety profile of lanreotide autogel. Lanreotide PRF was well tolerated and the pharmacokinetic profile suggests that a dosing interval of 12 weeks could be achievable. CLINICAL TRIAL REGISTRATION www.clinicaltrials.gov identifier is NCT02396953; EudraCT 2014-002389-62.
Collapse
Affiliation(s)
- Sebastian Neggers
- Medicine and Endocrinology, Erasmus University Medical Center, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Corin Badiu
- Department of Endocrinology National Institute of Endocrinology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Betina Biagetti
- Department of Endocrinology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | - Anne Petit
- Drug Product Development & Industrialization, Ipsen PharmSciences, Dreux, France
| | - Patrick Petrossians
- Department of Endocrinology, University Hospital of Liège, Sart Tilman Campus, Liège, Belgium
| | | | - David Rich
- Global Development, Rare Diseases, Ipsen BioInnovation, Abingdon, UK
| | - Zulfiya Shafigullina
- Department of Endocrinology, North-Western State Medical University Named after I.I Mechnikov, St. Petersburg, Russia
| | - Sergey Shustov
- Department of Endocrinology, North-Western State Medical University Named after I.I Mechnikov, St. Petersburg, Russia
| | - Anna Vydrych
- Department of Endocrinology, North-Western State Medical University Named after I.I Mechnikov, St. Petersburg, Russia
| |
Collapse
|
9
|
Cella D, Evans J, Feuilly M, Neggers S, Van Genechten D, Herman J, Khan MS. Patient and Healthcare Provider Perspectives of First-Generation Somatostatin Analogs in the Management of Neuroendocrine Tumors and Acromegaly: A Systematic Literature Review. Adv Ther 2021; 38:969-993. [PMID: 33432541 PMCID: PMC7799425 DOI: 10.1007/s12325-020-01600-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/05/2020] [Indexed: 11/24/2022]
Abstract
Introduction Somatostatin analogs (SSAs) are used to treat neuroendocrine tumors (NETs) and acromegaly. Two first-generation SSAs, octreotide long-acting release (OCT LAR) and lanreotide autogel/depot (LAN), are available. A systematic literature review (SLR) was conducted to investigate which characteristics beyond efficacy are most important in patient and healthcare practitioner (HCP) experience of LAN and OCT when used to treat acromegaly and NETs. Methods MEDLINE, Embase, the Cochrane Library, and Database of Abstracts of Reviews of Effect were searched from database inception to January 2019 with terms for first-generation SSAs, NETs, acromegaly, preferences, decision-making, and human factors. Key congresses in 2016–2018 and SLR bibliographies were hand-searched. Two independent reviewers screened articles at title/abstract and full-text stage. Publications fulfilling pre-specified inclusion criteria reported patient or HCP perspectives of LAN or OCT, or any factors affecting treatment perspectives for NETs or acromegaly. Results A total of 1110 unique records were screened, of which 21 studies were included, reporting from the perspectives of patients (n = 18) and/or HCPs (n = 9). Perspectives were collected using shared decision-making frameworks, questionnaires, informal patient opinion, and a Delphi panel. Where patient preference was specifically reported, LAN was preferred in 4/5 studies and OCT LAR in 1/5. Common factors underlying treatment experience included technical problems with injections and associated pain, emotional quality/anxiety of injections, time and convenience of treatment administration, and independence. Immediate aspects of injections appeared most important to patients, though the possibilities of extended dosing intervals and self-/partner-injection with LAN were also notable factors. Conclusions Study outcomes favored LAN in this SLR, with factors surrounding injection administration most influential in treatment experience. The findings of this SLR provide a basis that could inform development of decision-making criteria, with patient and HCP treatment perspectives considered. Future studies should utilize a common method to report preference and associated drivers. Supplementary Information The online version contains supplementary material available at 10.1007/s12325-020-01600-x.
Collapse
Affiliation(s)
- David Cella
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | | | | | - Sebastian Neggers
- Department of Medicine, Section Endocrinology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dirk Van Genechten
- Belgian Neuroendocrine Tumour (NET) and Multiple Endocrine Neoplasia (MEN) Association, Blankenberge, Belgium
- International Neuroendocrine Cancer Alliance (INCA), Boston, MA, USA
| | - Jackie Herman
- Canadian Neuroendocrine Tumour Society, Cornwall, ON, Canada
| | | |
Collapse
|
10
|
Álvarez-Escolá C, Venegas-Moreno EM, García-Arnés JA, Blanco-Carrera C, Marazuela-Azpiroz M, Gálvez-Moreno MÁ, Menéndez-Torre E, Aller-Pardo J, Salinas-Vert I, Resmini E, Torres-Vela EM, Gonzalo-Redondo MÁ, Vílchez-Joya R, de Miguel-Novoa MP, Halperín-Rabinovich I, Páramo-Fernández C, de la Cruz-Sugranyes G, Houchard A, Picó-Alfonso AM. ACROSTART: A retrospective study of the time to achieve hormonal control with lanreotide Autogel treatment in Spanish patients with acromegaly. ACTA ACUST UNITED AC 2019; 66:320-329. [PMID: 30773338 DOI: 10.1016/j.endinu.2018.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/28/2018] [Accepted: 12/06/2018] [Indexed: 10/27/2022]
Abstract
OBJECTIVES The ACROSTART study was intended to determine the time to achieve normalization of GH and IGF-I levels in responding patients with acromegaly administered different dosage regimens of lanreotide Autogel (Somatuline® Autogel®). METHODS From March 2013 to October 2013, clinical data from 57 patients from 17 Spanish hospitals with active acromegaly treated with lanreotide for ≥4 months who achieved hormonal control (GH levels <2.5ng/ml and/or normalized IGF-I levels in ≥2 measurements) were analyzed. The primary objective was to determine the time from start of lanreotide treatment to hormonal normalization. RESULTS Median patient age was 64 years, 21 patients were male, 39 patients had undergone surgery, and 14 patients had received radiotherapy. Median hormonal values at start of lanreotide treatment were: GH, 2.6ng/ml; IGF-I, 1.6×ULN. The most common starting dose of lanreotide was 120mg (29 patients). The main initial regimens were 60mg/4 weeks (n=13), 90mg/4 weeks (n=6), 120mg/4 weeks (n=13), 120mg/6 weeks (n=6), and 120mg/8 weeks (n=9). An initial treatment regimen with a long interval (≥6 weeks) was administered in 25 patients. Mean duration of lanreotide treatment was 68 months (7-205). Median time to achieve hormonal control was 4.9 months. Injections were managed without healthcare assistance in 13 patients. Median number of visits to endocrinologists until hormonal control was achieved was 3. Fifty-one patients were "satisfied"/"very satisfied" with treatment and 49 patients did not miss any dose. CONCLUSIONS Real-life treatment with lanreotide Autogel resulted in early hormonal control in responding patients, with high treatment adherence and satisfaction despite disparity in starting doses and dosing intervals.
Collapse
Affiliation(s)
| | | | | | - Concepción Blanco-Carrera
- Endocrinology Department, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Mónica Marazuela-Azpiroz
- Endocrinology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid, Instituto Princesa, Madrid, Spain
| | | | - Edelmiro Menéndez-Torre
- Endocrinology and Nutrition Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Javier Aller-Pardo
- Endocrinology Department, Neuroendocrinology & Endocrine Oncology Unit, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Isabel Salinas-Vert
- Endocrinology and Nutrition Department, Hospital Universitario Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Eugenia Resmini
- Hospital Sant Pau, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER, Unidad 747), IIB-Sant Pau, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | - Ricardo Vílchez-Joya
- Endocrinology and Nutrition Service, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - María Paz de Miguel-Novoa
- Endocrinology Department, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | | | - Aude Houchard
- Statistics Department, IPSEN PHARMA, Boulogne-Billancourt, France
| | | | | |
Collapse
|
11
|
Abreu C, Guinto G, Mercado M. Surgical-pharmacological interactions in the treatment of acromegaly. Expert Rev Endocrinol Metab 2019; 14:35-42. [PMID: 30595057 DOI: 10.1080/17446651.2019.1559729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/13/2018] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Acromegaly requires a multimodal treatment approach that includes surgery by an expert pituitary neurosurgeon, pharmacological treatment with one or more of the available drugs and radiation therapy. These treatment alternatives are not mutually exclusive but rather complement each other when properly indicated in the individual patient. In this review, we summarize and analyze the available data concerning the choice of the surgical approach (microscopy vs. endoscopy) and the interactions between medical treatment with somatostatin analogs and pituitary surgery. AREAS COVERED Technical aspects, complications and outcome of transsphenoidal surgery (TSS); Advantages and disadvantages of the microscopic and endoscopic approaches; Safety and efficacy of somatostatin analogs (SSA); Primary pharmacological therapy versus primary TSS; Benefits of the preoperative treatment with SSA; and the effect of surgical tumor debulking in the therapeutic response to SSA. EXPERT COMMENTARY Continuing efforts at improving surgical techniques and at generating more efficacious pharmacological therapies for acromegaly are likely to improve the outcome of these patients. However, an integral approach of the patient aimed not only at achieving biochemical criteria of cure but also at treating the individual comorbidities is mandatory to improve the quality of life of these patients and to reduce their mortality rate.
Collapse
Affiliation(s)
- Coralys Abreu
- a Endocrinology Service , Centro Medico Nacional 20 de Noviembre, ISSSTE , Mexico City , Mexico
| | - Gerardo Guinto
- b Neurological Center , American British Cowdray Medical Center , Mexico City , Mexico
| | - Moisés Mercado
- c Experimental Endocrinology Unit , Hospital de Especialidades, Centro Medico Nacional S. XXI, IMSS , Mexico City , Mexico
| |
Collapse
|
12
|
Godara A, Siddiqui NS, Byrne MM, Saif MW. The safety of lanreotide for neuroendocrine tumor. Expert Opin Drug Saf 2018; 18:1-10. [PMID: 30582380 DOI: 10.1080/14740338.2019.1559294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Lanreotide autogel is a synthetic somatostatin analogue which has been FDA and EMA approved for unresectable, well to moderately differentiated, locally advanced or metastatic gastroenteropancreatic neuroendocrine tumor. Its action is mediated by its affinity to somatostatin receptors, especially sst2 and sst5 receptors. Its longer half-life offers the convenience of 4-week dosing over the need for frequent injections of short-acting somatostatin analogues. Areas covered: Lanreotide ATG offers progression-free survival benefit in locally advanced or metastatic neuroendocrine tumor (NET) compared to placebo, reflecting a strong antiproliferative signal. As lanreotide is commonly used for management of NET, it is imperative to recognize and appropriately manage any drug-related toxicities. In this review, we will provide an overview of the toxicity with lanreotide and its management. Expert opinion: Lanreotide is highly effective in managing carcinoid symptoms and has a robust anti-tumor effect in NET. Overall, it is well tolerated with low rates of treatment discontinuation due to toxicity. It's toxicity profile is mostly predictable, and patients should be informed of the transient nature of some of the upfront toxicities.
Collapse
Affiliation(s)
- Amandeep Godara
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Nauman S Siddiqui
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Margaret M Byrne
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| | - Muhammad Wasif Saif
- a Gastrointestinal Oncology Program and Experimental therapeutics, Division of Hematology/Oncology , Tufts Medical Center - Tufts University School of Medicine , Boston , MA , USA
| |
Collapse
|
13
|
Abstract
Purpose Peptide drugs for antineoplastic therapies usually have low oral bioavailability and short in vivo half-lives, requiring less preferred delivery methods. Lanreotide depot is a sustained-release somatostatin analog (SSA) formulation produced via an innovative peptide self-assembly method. Lanreotide is approved in the USA and Europe to improve progression-free survival (PFS) in patients with unresectable gastroenteropancreatic neuroendocrine tumors (GEP-NETs) and also approved in Europe for symptom control in carcinoid syndrome associated with GEP-NETs. This review discusses how the distinct molecule and formulation of lanreotide depot provide advantages to patients and health care providers, as well as the most recent clinical evidence demonstrating the safety and efficacy of lanreotide depot in inhibiting tumor growth and controlling hormonal symptoms in GEP-NETs. Methodology and Results The lanreotide depot formulation confers a remarkable pharmacokinetic profile with no excipients, comprised only of lanreotide acetate and water. Of note, lanreotide depot constitutes an example for peptide self-assembly based formulations, providing insights that could help future development of sustained-release formulations of other antineoplastic peptides. Most patients with GEP-NETs will present with inoperable or incurable disease; thus, medical management for symptoms and tumor control plays a crucial role. Recent long-term clinical studies have demonstrated that lanreotide depot is well tolerated, prolongs PFS in GEP-NET patients, and significantly reduces symptoms related to carcinoid syndrome. Conclusions The unique depot formulation and delivery method of lanreotide confer advantages in the treatment of metastatic GEP-NETs, contributing to improvements in NET-related symptoms and PFS without reducing quality of life in this patient population.
Collapse
|
14
|
Population Pharmacokinetic Analysis of Lanreotide Autogel®/Depot in the Treatment of Neuroendocrine Tumors: Pooled Analysis of Four Clinical Trials. Clin Pharmacokinet 2015; 55:461-73. [DOI: 10.1007/s40262-015-0329-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
15
|
Neggers SJCMM, Pronin V, Balcere I, Lee MK, Rozhinskaya L, Bronstein MD, Gadelha MR, Maisonobe P, Sert C, van der Lely AJ. Lanreotide Autogel 120 mg at extended dosing intervals in patients with acromegaly biochemically controlled with octreotide LAR: the LEAD study. Eur J Endocrinol 2015; 173:313-23. [PMID: 26047625 PMCID: PMC4544680 DOI: 10.1530/eje-15-0215] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/05/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate extended dosing intervals (EDIs) with lanreotide Autogel 120 mg in patients with acromegaly previously biochemically controlled with octreotide LAR 10 or 20 mg. DESIGN AND METHODS Patients with acromegaly had received octreotide LAR 10 or 20 mg/4 weeks for ≥ 6 months and had normal IGF1 levels. Lanreotide Autogel 120 mg was administered every 6 weeks for 24 weeks (phase 1); depending on week-24 IGF1 levels, treatment was then administered every 4, 6 or 8 weeks for a further 24 weeks (phase 2). Hormone levels, patient-reported outcomes and adverse events were assessed. PRIMARY ENDPOINT proportion of patients on 6- or 8-week EDIs with normal IGF1 levels at week 48 (study end). RESULTS 107/124 patients completed the study (15 withdrew from phase 1 and two from phase 2). Of 124 patients enrolled, 77.4% were allocated to 6- or 8-week EDIs in phase 2 and 75.8% (95% CI: 68.3-83.3) had normal IGF1 levels at week 48 with the EDI (primary analysis). A total of 88.7% (83.1-94.3) had normal IGF1 levels after 24 weeks with 6-weekly dosing. GH levels were ≤ 2.5 μg/l in > 90% of patients after 24 and 48 weeks. Patient preferences for lanreotide Autogel 120 mg every 4, 6 or 8 weeks over octreotide LAR every 4 weeks were high. CONCLUSIONS Patients with acromegaly achieving biochemical control with octreotide LAR 10 or 20 mg/4 weeks are possible candidates for lanreotide Autogel 120 mg EDIs. EDIs are effective and well received among such patients.
Collapse
Affiliation(s)
- Sebastian J C M M Neggers
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Vyacheslav Pronin
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Inga Balcere
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Moon-Kyu Lee
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Liudmila Rozhinskaya
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Marcello D Bronstein
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Mônica R Gadelha
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Pascal Maisonobe
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Caroline Sert
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| | - Aart Jan van der Lely
- Department of EndocrinologyErasmus Medical Center, Rotterdam, NetherlandsDepartment of EndocrinologyI.M. Sechenov First Moscow State Medical University, Moscow, Russian FederationDepartment of EndocrinologyPauls Stradins Clinical University Hospital, Riga, LatviaDivision of Endocrinology and MetabolismSamsung Medical Center, Sungkyunkwan University, Seoul, Republic of KoreaDepartment of Neuroendocrinology and Bone DiseasesNational Endocrinology Research Centre, Moscow, Russian FederationNeuroendocrine UnitDivision of Endocrinology and Metabolism, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, BrazilEndocrine SectionHospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BrazilIpsen Boulogne-BillancourtFrance
| |
Collapse
|
16
|
Kyriakakis N, Chau V, Lynch J, Orme SM, Murray RD. Lanreotide autogel in acromegaly - a decade on. Expert Opin Pharmacother 2014; 15:2681-92. [PMID: 25307803 DOI: 10.1517/14656566.2014.970173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The novel formulation of lanreotide, lanreotide (LAN) autogel (ATG), has been available in Europe since 2001 and USA from 2006 for the treatment of acromegaly. It is one of only two clinically available somatostatin analogs available for use in acromegaly. Data relating to the use of ATG in acromegaly, specifically relating to comparison to octreotide (OCT) LAR and patient acceptability and preference, have been slow to accumulate. AREAS COVERED We performed a comprehensive review of the original literature relating to development, pharmacokinetics, acceptability and clinical efficacy of ATG. EXPERT OPINION LAN ATG is a novel formulation of LAN consequent on self-assembly of nanotubules in water. Diffusion between molecules within the nanotubules and surrounding tissue fluid in vivo leads to pseudo first-order pharmacokinetics. Efficacy is equivalent to the alternate long-acting somatostatin analog, OCT LAR, normalizing growth hormone and IGF-I levels in around 60 and 50% respectively. Control of tumor growth is observed in over 95% of patients, with 64% seeing a clinically significant reduction in tumor size. ATG is provided in a prefilled syringe for deep subcutaneous injection, allowing self-injection, and may be administered up to 8 weeks greatly improving convenience for the patient. The data strongly support consideration of ATG as the medical therapy of choice for patients with acromegaly.
Collapse
Affiliation(s)
- Nikolaos Kyriakakis
- St James's University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds Centre for Diabetes and Endocrinology, Department of Endocrinology , Beckett Street, Leeds LS9 7TF , UK +44 0 113 206 4578 ; +44 0 113 206 5065 ;
| | | | | | | | | |
Collapse
|
17
|
Burness CB, Dhillon S, Keam SJ. Lanreotide Autogel®: A Review of its Use in the Treatment of Patients with Acromegaly. Drugs 2014; 74:1673-91. [DOI: 10.1007/s40265-014-0283-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
18
|
Sowiński J, Sawicka N, Piątek K, Zybek A, Ruchała M. Pharmacoeconomic aspects of the treatment of pituitary gland tumours. Contemp Oncol (Pozn) 2013; 17:137-43. [PMID: 23788980 PMCID: PMC3685378 DOI: 10.5114/wo.2013.34616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 04/14/2013] [Accepted: 04/18/2013] [Indexed: 12/17/2022] Open
Abstract
Nowadays physicians are under economic pressure; therefore therapeutic decisions based on safety, efficacy, and the effectiveness of the medication also require economic analysis. The aim of this review is to discuss data concerning the cost-effectiveness of drug therapy in patients with hormonally active pituitary adenomas, namely growth hormone, adrenocorticotropic hormone, thyroid-stimulating hormone-secreting pituitary adenomas, prolactinoma and pituitary incidentaloma. In acromegalic patients using lanreotide is cheaper for health care payers and more convenient for physicians and patients because of the opportunity for self/partner injections, lower clogging risk and possibility of longer intervals between injections, while the efficacy is comparable with octreotide. Patients with prolactinomas should be treated with novel dopamine agonists, such as cabergoline or quinagolide, however, bromocriptine still remains a cheaper and almost as effective alternative. There are no easy methods or algorithms, but in general, extracting the maximum value from the investment in treatment is essential.
Collapse
Affiliation(s)
- Jerzy Sowiński
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poland
| | | | | | | | | |
Collapse
|
19
|
Garrido MJ, Cendrós JM, Ramis J, Peraire C, Obach R, Trocóniz IF. Pharmacodynamic Modeling of the Effects of Lanreotide Autogel on Growth Hormone and Insulin-Like Growth Factor 1. J Clin Pharmacol 2013; 52:487-98. [DOI: 10.1177/0091270011399761] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
20
|
Roemmler J, Schopohl J. Clinical experience with lanreotide for the treatment of acromegaly. Expert Rev Endocrinol Metab 2012; 7:139-149. [PMID: 30764005 DOI: 10.1586/eem.11.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Acromegaly is a rare disease, characterized in adults by its distinctive appearance of facial dysmorphism and swollen fingers. It is caused by an overproduction of growth hormone (GH) in more than 99% of patients and in nearly all cases is due to a pituitary adenoma. If surgical resection of the adenoma is not effective, medical treatment is usually the next treatment option. The most commonly used medications are the somatostatin analogues octreotide and lanreotide. Lanreotide is a synthetic somatostatin analogue and is available as slow-release microparticle (every 7-14 days) and prolonged-release liquid (autogel, every 28-56 days) formulations. Lanreotide autogel is a supersaturated aqueous formulation for deep subcutaneous injection and is sold in a ready-to-use prefilled syringe. This ease of use allows self or partner administration at home. This article reviews the use of lanreotide in the treatment of acromegaly and its advantages and disadvantages compared with other somatostatin analogues.
Collapse
Affiliation(s)
- Josefine Roemmler
- b Medizinische Klinik und Poliklinik IV, University of Munich, Ziemssenstr. 1, 80336 München, Germany.
| | - Jochen Schopohl
- a Medizinische Klinik und Poliklinik IV, University of Munich, Ziemssenstr. 1, 80336 München, Germany
| |
Collapse
|
21
|
Carmichael JD. Lanreotide depot deep subcutaneous injection: a new method of delivery and its associated benefits. Patient Prefer Adherence 2012; 6:73-82. [PMID: 22298946 PMCID: PMC3269320 DOI: 10.2147/ppa.s20783] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acromegaly is a rare disease characterized by excessive growth hormone secretion, usually from a pituitary tumor. Treatment options include surgery, medical therapy, and in some cases, radiation therapy. Current medical therapy consists of treatment with somatostatin analog medications or a growth hormone receptor antagonist. There are two somatostatin analogs currently in use, octreotide and lanreotide. Both are supplied in long-acting formulations and are of comparable biochemical efficacy. Lanreotide is supplied in a prefilled syringe and is injected into deep subcutaneous tissue. Studies have been conducted to assess the efficacy of self- or partner administration, and have demonstrated that injection of lanreotide can be accomplished reliably and safely outside a physician's office. For patients who have achieved biochemical control with lanreotide, the FDA has recently approved an extended dosing interval. Selected patients may be able to receive the medication less frequently with injections of 120 mg administered every 6 or 8 weeks. This review focuses on the use of lanreotide in the treatment of acromegaly, the safety and efficacy of the drug, and the benefits afforded to patients because of unique aspects of the delivery of lanreotide.
Collapse
Affiliation(s)
- John D Carmichael
- Correspondence: John D Carmichael, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA, Tel +1 310 423 2830, Fax +1 310 423 2819, Email
| |
Collapse
|
22
|
Zhang S, Shi R, Li C, Parivar K, Wang DD. Fixed dosing versus body size-based dosing of therapeutic peptides and proteins in adults. J Clin Pharmacol 2011; 52:18-28. [PMID: 21233304 DOI: 10.1177/0091270010388648] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Therapeutic biologics are often administered based on body size. A previous study has found that fixed dosing performs similarly to body size-based dosing in reducing intersubject variability in drug exposure across the mAbs studied. This study extended this evaluation to other therapeutic proteins and peptides. Eighteen therapeutic proteins and peptides with published population pharmacokinetic (PK) and/or pharmacodynamic (PD) models were selected for dosing approach evaluation. The relationships between body size and drug exposure (and PD end point when available) were evaluated, and simulation studies were conducted to compare the performance of the 2 dosing approaches. The results showed that fixed dosing performed better for 12 of 18 selected biologics in terms of reducing intersubject variability in exposure at both population and individual levels, whereas body size-based dosing performed better for the other 6 molecules. This result is consistent with the findings for mAbs. Therefore, fixed dosing is recommended for first-in-human studies of proteins and peptides along with mAbs. The final dosing approach for phase 3 studies should be determined based on a full assessment of body size effect on PK/PD when data are available and the therapeutic window of the drug.
Collapse
|
23
|
|
24
|
Castinetti F, Saveanu A, Morange I, Brue T. Lanreotide for the treatment of acromegaly. Adv Ther 2009; 26:600-12. [PMID: 19533047 DOI: 10.1007/s12325-009-0035-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Indexed: 01/12/2023]
Abstract
Lanreotide is an eight-amino acid peptide, which is an analog of the native somatostatin peptide, physiological inhibitor of growth hormone (GH). The drug shows high binding affinity for somatostatin receptors, SSTR2 and SSTR5, which is the primary mechanism considered to be responsible for decreasing GH secretion and GH cell proliferation in acromegaly. Two different formulations of lanreotide are currently available: lanreotide slow release, which requires intramuscular injection every 7-14 days, and lanreotide autogel, which requires deep subcutaneous injection every 4-8 weeks. Several studies have been published to date on the use of lanreotide in acromegaly. Antisecretory efficacy has been reported in 35%-70% of cases; this huge variability is probably explained by different indications (eg, primary or adjunctive postsurgical treatment), or the fact that some studies were based on patients known to be responders to somatostatin analogs. As a primary treatment, antisecretory efficacy was very similar, confirming the possibility of lanreotide as an option in cases of unsuccessful surgery, contraindication, or surgery refusal. Lanreotide also has antitumoral effects as it induces a decrease in tumor volume of [Symbol: see text]25% in 30%-70% of patients. This could be beneficial before transsphenoidal surgery, as a pretreatment, to decrease tumor volume and ease surgery; however, to date, advantages in terms of final remission or uncured status remain a matter of debate. Side effects are rare; the most frequent being gastrointestinal discomfort and increased risk of gallstone formation, and glucose metabolism modifications. Comparison with the other somatostatin analog, octreotide, tends to show identical levels of efficacy between both drugs. Lanreotide thus seems to be an effective treatment in acromegaly. To date, however, lanreotide is still considered as only suspending GH secretion, thus requiring prolonged and costly treatment.
Collapse
Affiliation(s)
- F Castinetti
- Department of Endocrinology, Université de la Méditerranée, France
| | | | | | | |
Collapse
|