1
|
Selaya SD, Abrigo N, Brown DG, Desai S, Beekman C, Faustino P, Shakleya D. A headspace GC-MS method to quantify nitrosamine impurities and precursors in drug products: Method validation and product testing. Biomed Chromatogr 2024:e6040. [PMID: 39510975 DOI: 10.1002/bmc.6040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
Pharmaceutical manufacturers are working to mitigate the formation of nitrosamine impurities in drug products. The work herein describes the development and validation of a headspace GC-MS method according to ICH Q2(R1) guidelines for the detection and quantification of NDMA, NDEA, NDIPA, and NEIPA in drug products. The analytical procedure was further modified to include detection and quantitation of DMF due to the potential decomposition pathway of DMF to form dimethylamine, a known precursor for NDMA formation. The NDMA impurity was detected in the "sartan" class of drug products between 0.1 and 113 ppm. The validated analytical procedure was applied in an investigation of approaches to mitigate nitrosamine formation in metformin drug products. The developed analytical procedures provide another tool for pharmaceutical manufacturers to evaluate drug products for nitrosamine impurities.
Collapse
Affiliation(s)
- Susan Daniela Selaya
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
- Office of Product Quality Assessment, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
| | - Nicolas Abrigo
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
- Office of Product Quality Assessment, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
| | - Dustin G Brown
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
| | - Saaniya Desai
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
| | - Christopher Beekman
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
- Office of Regulatory Science, Center for Food Safety and Applied Nutrition, College Park, Maryland, USA
| | - Patrick Faustino
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
| | - Diaa Shakleya
- Office of Pharmaceutical Quality Research, Center for Drug Evaluation and Research Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
2
|
Nogueira RC, Sanches-Lopes JM, Oliveira-Paula GH, Tanus-Santos JE. Inhibitors of gastric acid secretion increase oxidative stress and matrix metalloproteinase-2 activity leading to vascular remodeling. Mol Cell Biochem 2024; 479:3141-3152. [PMID: 38302836 DOI: 10.1007/s11010-023-04921-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/20/2023] [Indexed: 02/03/2024]
Abstract
The use of inhibitors of gastric acid secretion (IGAS), especially proton pump inhibitors (PPI), has been associated with increased cardiovascular risk. While the mechanisms involved are not known, there is evidence supporting increased oxidative stress, a major activator of matrix metalloproteinases (MMP), as an important player in such effect. However, there is no study showing whether other IGAS such as histamine H2-receptor blockers (H2RB) cause similar effects. This study aimed at examining whether treatment with the H2RB ranitidine promotes oxidative stress resulting in vascular MMP activation and corresponding functional and structural alterations in the vasculature, as compared with those found with the PPI omeprazole. Male Wistar rats were treated (4 weeks) with vehicle (2% tween 20), omeprazole (10 mg/Kg/day; i.p.) or ranitidine (100 mg/Kg/day; gavage). Then the aorta was collected to perform functional, biochemical, and morphometric analysis. Both ranitidine and omeprazole increased gastric pH and oxidative stress assessed in situ with the fluorescent dye dihydroethidium (DHE) and with lucigenin chemiluminescence assay. Both IGAS augmented vascular activated MMP-2. These findings were associated with aortic remodeling (increased media/lumen ratio and number of cells/μm2). Both IGAS also impaired the endothelium-dependent relaxation induced by acetylcholine (isolated aortic ring preparation). This study provides evidence that the H2RB ranitidine induces vascular dysfunction, redox alterations, and remodeling similar to those found with the PPI omeprazole. These findings strongly suggest that IGAS increase oxidative stress and matrix metalloproteinase-2 activity leading to vascular remodeling, which helps to explain the increased cardiovascular risk associated with the use of those drugs.
Collapse
Affiliation(s)
- Renato C Nogueira
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, 14049-900, Brazil
| | - Jéssica M Sanches-Lopes
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, 14049-900, Brazil
| | - Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, 14049-900, Brazil
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, 14049-900, Brazil.
| |
Collapse
|
3
|
Shinde S, Croft KD, Hodgson JM, Bondonno CP. Rapid analysis of N-nitrosamines in urine using ultra high-pressure liquid chromatography-mass spectrometry. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024. [PMID: 39469813 DOI: 10.1039/d4ay01870b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
N-Nitrosamines, carcinogenic compounds present in dietary and environmental sources and formed endogenously, are believed to be linked with the presence of nitrate and nitrite, both within dietary sources and after intake. To fully evaluate this potential threat to human health, an accurate analytical method to measure N-nitrosamines in biological matrices is necessary. We report a simple, fast, selective mass spectrometry method to detect N-nitrosamines in human urine. Analysis of seven N-nitrosamines, N-nitrosodimethylamine (NDMA), N-nitrosomethylethylamine (NMEA), N-nitrosodiethylamine (NDEA), N-nitrosopiperdine (NPIP), N-nitrosopyrrolidine (NPYR), N-nitrosodi-N-propylamine (NDPA) and N-nitrosodi-N-butylamine (NDBA) in urine was quantitated using Ultra High-Pressure Liquid Chromatography-tandem Mass spectrometry (UHPLC-MS/MS). A Sorbent supported Liquid Extraction (SLE) method was employed to extract N-nitrosamines from 24 hour collected human urine samples. The percent recovery varied between 74.3 to 110 and the limit of detection and limit of quantification ranged from 0.1 to 0.85 ng mL-1 and 0.22 to 2.06 ng mL-1 respectively. Precision for inter-day and intra-day assay yielded a % coefficient of variation between 4-10% for all measured compounds in urine. Linear regression analysis of calibration curves for N-nitrosamines measured in urine in the concentration range 0.4-12.8 ng mL-1 gave correlation coefficients, R2 0.9874-0.9962. Urinary excretion of N-nitrosamines measured in ten healthy subjects resulted in detection of most of the N-nitrosamines including NDMA, NDEA, NPYR, NDPA and NDBA by this method.
Collapse
Affiliation(s)
- S Shinde
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - K D Croft
- School of Biomedical Science, University of Western Australia, Perth, Western Australia, Australia
| | - J M Hodgson
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Level 3, Royal Perth Hospital Research Foundation. Rear 50 Murray St, Joondalup, Perth, Western Australia, Australia WA 6000.
| | - C P Bondonno
- Medical School, University of Western Australia, Perth, Western Australia, Australia
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Level 3, Royal Perth Hospital Research Foundation. Rear 50 Murray St, Joondalup, Perth, Western Australia, Australia WA 6000.
| |
Collapse
|
4
|
Paustenbach DJ, Brown SE, Heywood JJ, Donnell MT, Eaton DL. Risk characterization of N-nitrosodimethylamine in pharmaceuticals. Food Chem Toxicol 2024; 186:114498. [PMID: 38341171 DOI: 10.1016/j.fct.2024.114498] [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: 09/28/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Since 2018, N-nitrosodimethylamine (NDMA) has been a reported contaminant in numerous pharmaceutical products. To guide the pharmaceutical industry, FDA identified an acceptable intake (AI) of 96 ng/day NDMA. The approach assumed a linear extrapolation from the Carcinogenic Potency Database (CPDB) harmonic-mean TD50 identified in chronic studies in rats. Although NDMA has been thought to act as a mutagenic carcinogen in experimental animals, it has not been classified as a known human carcinogen by any regulatory agency. Humans are exposed to high daily exogenous and endogenous doses of NDMA. Due to the likelihood of a threshold dose for NDMA-related tumors in animals, we believe that there is ample scientific basis to utilize the threshold-based benchmark dose or point-of-departure (POD) approach when estimating a Permissible Daily Exposure limit (PDE) for NDMA. We estimated that 29,000 ng/kg/day was an appropriate POD for calculating a PDE. Assuming an average bodyweight of 50 kg, we expect that human exposures to NDMA at doses below 5800 ng/day in pharmaceuticals would not result in an increased risk of liver cancer, and that there is little, if any, risk for any other type of cancer, when accounting for the mode-of-action in humans.
Collapse
Affiliation(s)
- D J Paustenbach
- Paustenbach and Associates, 970 West Broadway, Suite E, Jackson, WY, USA
| | - S E Brown
- Paustenbach and Associates, 207 Canyon Blvd, Boulder, CO, USA.
| | - J J Heywood
- Paustenbach and Associates, 207 Canyon Blvd, Boulder, CO, USA
| | - M T Donnell
- Valeo Sciences LLC, 333 Corporate Drive, Suite 130, Ladera Ranch, CA, USA
| | - D L Eaton
- Professor Emeritus, Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
5
|
Shakleya D, Asmelash B, Alayoubi A, Abrigo N, Mohammad A, Wang J, Zhang J, Yang J, Marzan TA, Li D, Shaklah M, Alsharif FM, Desai S, Faustino PJ, Ashraf M, O'Connor T, Vera M, Raw A, Sayeed VA, Keire D. Bumetanide as a Model NDSRI Substrate: N-nitrosobumetanide Impurity Formation and its Inhibition in Bumetanide Tablets. J Pharm Sci 2023; 112:3075-3087. [PMID: 37364772 DOI: 10.1016/j.xphs.2023.06.013] [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: 05/16/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023]
Abstract
Nitrosamine compounds are classified as potential human carcinogens, the origin of these impurities can be broadly classified in two categories, nitrosamine impurity found in drug products that are not associated with the Active Pharmaceutical Ingredient (API), such as N-nitrosodimethylamine (NDMA) or nitrosamine impurities associated with the API, such as nitrosamine drug substance-related impurities (NDSRIs). The mechanistic pathway for the formation of these two classes of impurities can be different and the approach to mitigate the risk should be tailored to address the specific concern. In the last couple of years number of NDSRIs have been reported for different drug products. Though, not the only contributing factor for the formation of NDSIRs, it is widely accepted that the presence of residual a nitrites/nitrates in the components used in the manufacturing of the drug products can be the primary contributor to the formation of NDSRIs. Approaches to mitigate the formation of NDSRIs in drug products include the use of antioxidants or pH modifiers in the formulation. The primary objective of this work was to evaluate the role of different inhibitors (antioxidants) and pH modifiers in tablet formulations prepared in-house using bumetanide (BMT) as a model drug to mitigate the formation of N-nitrosobumetanide (NBMT). A multi-factor study design was created, and several bumetanide formulations were prepared by wet granulation with and without sodium nitrite spike (100 ppm) and different antioxidants (ascorbic acid, ferulic acid or caffeic acid) at three concentrations (0.1%, 0.5% or 1% of the total tablet weight). Formulations with acidic and basic pH were also prepared using 0.1 N hydrochloric acid and 0.1 N sodium bicarbonate, respectively. The formulations were subjected to different storage (temperature and humidity) conditions over 6 months and stability data was collected. The rank order of N-nitrosobumetanide inhibition was highest with alkaline pH formulations, followed by formulations with ascorbic acid, caffeic acid or ferulic acid present. In summary, we hypothesize that maintaining a basic pH or the addition of an antioxidant in the drug product can mitigate the conversion of nitrite to nitrosating agent and thus reduce the formation of bumetanide nitrosamines.
Collapse
Affiliation(s)
- Diaa Shakleya
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| | - Bethel Asmelash
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of lifecycle Drug product, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Alaadin Alayoubi
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Nicolas Abrigo
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Adil Mohammad
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Jiang Wang
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Jinhui Zhang
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Jingyue Yang
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Tim Andres Marzan
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - David Li
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Maha Shaklah
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Fahd M Alsharif
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Saaniya Desai
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Patrick J Faustino
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Muhammad Ashraf
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Thomas O'Connor
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Matthew Vera
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of lifecycle Drug product, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Andre Raw
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of lifecycle Drug product, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - Vilayat A Sayeed
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of lifecycle Drug product, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - David Keire
- Food and Drug Administration, Center for Drug Evaluation and Research, Office of Pharmaceutical Quality, Office of Testing and Research, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| |
Collapse
|
6
|
You SC, Seo SI, Falconer T, Yanover C, Duarte-Salles T, Seager S, Posada JD, Shah NH, Nguyen PA, Kim Y, Hsu JC, Van Zandt M, Hsu MH, Lee HL, Ko H, Shin WG, Pratt N, Park RW, Reich CG, Suchard MA, Hripcsak G, Park CH, Prieto-Alhambra D. Ranitidine Use and Incident Cancer in a Multinational Cohort. JAMA Netw Open 2023; 6:e2333495. [PMID: 37725377 PMCID: PMC10509724 DOI: 10.1001/jamanetworkopen.2023.33495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/02/2023] [Indexed: 09/21/2023] Open
Abstract
Importance Ranitidine, the most widely used histamine-2 receptor antagonist (H2RA), was withdrawn because of N-nitrosodimethylamine impurity in 2020. Given the worldwide exposure to this drug, the potential risk of cancer development associated with the intake of known carcinogens is an important epidemiological concern. Objective To examine the comparative risk of cancer associated with the use of ranitidine vs other H2RAs. Design, Setting, and Participants This new-user active comparator international network cohort study was conducted using 3 health claims and 9 electronic health record databases from the US, the United Kingdom, Germany, Spain, France, South Korea, and Taiwan. Large-scale propensity score (PS) matching was used to minimize confounding of the observed covariates with negative control outcomes. Empirical calibration was performed to account for unobserved confounding. All databases were mapped to a common data model. Database-specific estimates were combined using random-effects meta-analysis. Participants included individuals aged at least 20 years with no history of cancer who used H2RAs for more than 30 days from January 1986 to December 2020, with a 1-year washout period. Data were analyzed from April to September 2021. Exposure The main exposure was use of ranitidine vs other H2RAs (famotidine, lafutidine, nizatidine, and roxatidine). Main Outcomes and Measures The primary outcome was incidence of any cancer, except nonmelanoma skin cancer. Secondary outcomes included all cancer except thyroid cancer, 16 cancer subtypes, and all-cause mortality. Results Among 1 183 999 individuals in 11 databases, 909 168 individuals (mean age, 56.1 years; 507 316 [55.8%] women) were identified as new users of ranitidine, and 274 831 individuals (mean age, 58.0 years; 145 935 [53.1%] women) were identified as new users of other H2RAs. Crude incidence rates of cancer were 14.30 events per 1000 person-years (PYs) in ranitidine users and 15.03 events per 1000 PYs among other H2RA users. After PS matching, cancer risk was similar in ranitidine compared with other H2RA users (incidence, 15.92 events per 1000 PYs vs 15.65 events per 1000 PYs; calibrated meta-analytic hazard ratio, 1.04; 95% CI, 0.97-1.12). No significant associations were found between ranitidine use and any secondary outcomes after calibration. Conclusions and Relevance In this cohort study, ranitidine use was not associated with an increased risk of cancer compared with the use of other H2RAs. Further research is needed on the long-term association of ranitidine with cancer development.
Collapse
Affiliation(s)
- Seng Chan You
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea
- Institute for Innovation in Digital Healthcare, Yonsei University, Seoul, Korea
| | - Seung In Seo
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Thomas Falconer
- Department of Biomedical Informatics, Columbia University, New York, New York
| | | | - Talita Duarte-Salles
- Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina, Barcelona, Spain
| | | | - Jose D. Posada
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Nigam H. Shah
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Phung-Anh Nguyen
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taiwan
| | - Yeesuk Kim
- Department of Orthopaedic Surgery, College of Medicine, Hanyang University, Seoul, Korea
| | - Jason C. Hsu
- International PhD Program in Biotech and Healthcare Management, College of Management, Taipei Medical University, Taipei, Taiwan
| | | | - Min-Huei Hsu
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taiwan
| | - Hang Lak Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Heejoo Ko
- College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woon Geon Shin
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Nicole Pratt
- Quality Use of Medicines and Pharmacy Research Centre, Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Rae Woong Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Gyeonggi-do, Korea
| | | | - Marc A. Suchard
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles
- VA Informatics and Computing Infrastructure, US Department of Veterans Affairs, Salt Lake City, Utah
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University, New York, New York
- Medical Informatics Services, New York-Presbyterian Hospital, New York, New York
| | - Chan Hyuk Park
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Daniel Prieto-Alhambra
- Pharmaco- and Device Epidemiology, Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
- Department of Medical Informatics, Erasmus Medical Center University, Rotterdam, Netherlands
| |
Collapse
|
7
|
Kang H, Nam CM, Choi DW, Park S. Effects of ranitidine and nizatidine on the risk of gastrointestinal cancer. Front Oncol 2023; 13:1182174. [PMID: 37576886 PMCID: PMC10415903 DOI: 10.3389/fonc.2023.1182174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Purpose Gastrointestinal (GI) cancer occurs in digestive organs such as the stomach, colon, liver, esophagus, and pancreas. About 83,034 cases occurred in Korea alone in 2020. Dietary factors, alcohol consumption, Helicobacter pylori (H. pylori), and lifestyle factors increase the incidence of diseases such as gastritis, peptic ulcer, pancreatitis, and gastroesophageal reflux disease (GERD), which can develop into GI cancer. However, in 2019, the US Food and Drug Administration announced that the drugs ranitidine and nizatidine, which are used for digestive disorders, contain carcinogens. In this study, we investigated the effects of ranitidine and nizatidine on the development of GI cancer. Materials and methods In this study, using National Health Insurance Service-National Sample Cohort (NHIS-NSC) version 2.5 (updated from 2002 to 2019), subjects who developed GI cancer were enrolled in the case group, and those who were at risk of, but did not develop, cancer were enrolled in the control group. Thereafter, risk-set matching was performed (1:3 ratio) by sex and age at the time of diagnosis of cancer in the case group. Through this procedure, 22,931 cases and 68,793 controls were identified. The associations of ranitidine and/or nizatidine with GI cancer were confirmed by adjusted odds ratios (aORs) and 95% confidence intervals (CIs) calculated through conditional logistic regression analysis. Results The aORs of ranitidine and/or nizatidine users were lower than those of nonusers in all average prescription days groups (< 30 days/year: aOR [95% CI] = 0.79 [0.75-0.82]; 30-59 days/year: aOR [95% CI] = 0.66 [0.59-0.73]; 60-89 days/year: aOR [95% CI] = 0.69 [0.59-0.81]; ≥ 90 days/year: aOR [95% CI] = 0.69 [0.59-0.79]). Sensitivity analyses were conducted with different lag periods for the onset of GI cancer after drug administration, and these analyses yielded consistent results. Additional analyses were also performed by dividing subjects into groups based on cancer types and CCI scores, and these analyses produced the same results. Conclusion Our study, using nationwide retrospective cohort data, did not find evidence suggesting that ranitidine and nizatidine increase the risk of GI cancer. In fact, we observed that the incidence of GI cancer was lower in individuals who used the drugs compared to nonusers. These findings suggest a potential beneficial effect of these drugs on cancer risk, likely attributed to their ability to improve digestive function.
Collapse
Affiliation(s)
- Hyejung Kang
- Department of Health Informatics and Biostatistics, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Chung Mo Nam
- Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong-Woo Choi
- Cancer Big Data Center, National Cancer Control Institute, National Cancer Center, Goyang, Republic of Korea
| | - Sohee Park
- Department of Health Informatics and Biostatistics, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
8
|
De Palma R, Patel V, Florian J, Keire D, Selaya D, Strauss DG, Rouse R, Matta MK. A Bioanalytical Method for Quantification of N-nitrosodimethylamine (NDMA) in Human Plasma and Urine with Different Meals and following Administration of Ranitidine. J Pharm Sci 2023; 112:1315-1323. [PMID: 36736776 DOI: 10.1016/j.xphs.2023.01.026] [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: 12/01/2022] [Revised: 01/13/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
Control of N-nitrosoamine impurities is important for ensuring the safety of drug products. Findings of nitrosamine impurities in some drug products led FDA to develop new guidance providing recommendations for manufacturers towards prevention and detection of nitrosamine impurities in pharmaceutical products. One of these products, ranitidine, also had a published in vivo study, which has since been retracted by its authors, suggesting a potential for in vivo conversion of ranitidine to the probable human carcinogen, N-nitrosodimethylamine (NDMA). FDA subsequently initiated a randomized, double-blind, placebo-controlled, crossover clinical investigation to assess the potential for in vivo conversion of ranitidine to NDMA with different meals. A bioanalytical method toward characterization of NDMA formation was needed as previously published methods did not address potential NDMA formation after biofluid collection. Therefore, a bioanalytical method was developed and validated as per FDA's Bioanalytical Method Validation guidance. An appropriate surrogate matrix for calibration standards and quality control sample preparation for both liquid matrices (human plasma and urine) was optimized to minimize the artifacts of assay measurements and monitor basal NDMA levels. Interconversion potential of ranitidine to NDMA was monitored during method validation by incorporating the appropriate quality control samples. The validated methods for NDMA were linear from 15.6 pg/mL to 2000 pg/mL. Low sample volumes (2 mL for urine and 1 mL for plasma) made this method suitable for clinical study samples and helped to evaluate the influence of ranitidine administration and meal types on urinary excretion of NDMA in human subjects.
Collapse
Affiliation(s)
- Ryan De Palma
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States
| | - Vikram Patel
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States
| | - Jeffry Florian
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States
| | - David Keire
- Office of Testing and Research, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States
| | - Daniela Selaya
- Office of Testing and Research, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States
| | - David G Strauss
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States
| | - Rodney Rouse
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States
| | - Murali K Matta
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drugs Evaluation and Research, US Food and Drug Administration, United States.
| |
Collapse
|
9
|
Chiu K, Racz R, Burkhart K, Florian J, Ford K, Iveth Garcia M, Geiger RM, Howard KE, Hyland PL, Ismaiel OA, Kruhlak NL, Li Z, Matta MK, Prentice KW, Shah A, Stavitskaya L, Volpe DA, Weaver JL, Wu WW, Rouse R, Strauss DG. New science, drug regulation, and emergent public health issues: The work of FDA's division of applied regulatory science. Front Med (Lausanne) 2023; 9:1109541. [PMID: 36743666 PMCID: PMC9893027 DOI: 10.3389/fmed.2022.1109541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/13/2022] [Indexed: 01/20/2023] Open
Abstract
The U.S. Food and Drug Administration (FDA) Division of Applied Regulatory Science (DARS) moves new science into the drug review process and addresses emergent regulatory and public health questions for the Agency. By forming interdisciplinary teams, DARS conducts mission-critical research to provide answers to scientific questions and solutions to regulatory challenges. Staffed by experts across the translational research spectrum, DARS forms synergies by pulling together scientists and experts from diverse backgrounds to collaborate in tackling some of the most complex challenges facing FDA. This includes (but is not limited to) assessing the systemic absorption of sunscreens, evaluating whether certain drugs can convert to carcinogens in people, studying drug interactions with opioids, optimizing opioid antagonist dosing in community settings, removing barriers to biosimilar and generic drug development, and advancing therapeutic development for rare diseases. FDA tasks DARS with wide ranging issues that encompass regulatory science; DARS, in turn, helps the Agency solve these challenges. The impact of DARS research is felt by patients, the pharmaceutical industry, and fellow regulators. This article reviews applied research projects and initiatives led by DARS and conducts a deeper dive into select examples illustrating the impactful work of the Division.
Collapse
Affiliation(s)
- Kimberly Chiu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Rebecca Racz
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Keith Burkhart
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Jeffry Florian
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Kevin Ford
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - M. Iveth Garcia
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Robert M. Geiger
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Kristina E. Howard
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Paula L. Hyland
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Omnia A. Ismaiel
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Naomi L. Kruhlak
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Zhihua Li
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Murali K. Matta
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Kristin W. Prentice
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States,Booz Allen Hamilton, McLean, VA, United States
| | - Aanchal Shah
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States,Booz Allen Hamilton, McLean, VA, United States
| | - Lidiya Stavitskaya
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Donna A. Volpe
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - James L. Weaver
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Wendy W. Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - Rodney Rouse
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States
| | - David G. Strauss
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, United States,*Correspondence: David G. Strauss,
| |
Collapse
|
10
|
Zhou A, Chen X, Li C, Yang W, He J, Fang T, Chen W, Xu Y, Ge H, Chen Z, Ning X. Orthogonal Chemical Reporter Strategy Enables Sensitive and Specific SERS Detection of Hydrazine Derivatives. ACS APPLIED MATERIALS & INTERFACES 2023; 15:2054-2066. [PMID: 36579636 DOI: 10.1021/acsami.2c16982] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Hydrazine and its derivatives are well-known environmental hazards and biological carcinogens; therefore, there is a great need for a powerful workflow solution for protecting the public from unexpected exposure to toxic contaminants. Recently, functional surface-enhanced Raman scattering (SERS) exhibits enormous benefits in sensing trace biochemical substances due to its fingerprint-like identification of individual molecules, making it an ideal method for detecting and quantifying hydrazine. Herein, for the first time, we integrated the orthogonal chemical reporter strategy with SERS to build an intelligent hydrazine detection platform (orthogonal chemical SERS, ocSERS), in which 4-mercaptobenzaldehyde was incorporated on a nanoimprinted gold nanopillar array, which acted as an orthogonal coupling partner of hydrazine to form Raman active benzaldehyde hydrazone, allowing for sensitively detecting hydrazine with a detection limit of 10-13 M in complex circumstances. Particularly, ocSERS could effectively identify the carcinogen N-nitrosodimethylamine (NDMA) after its reduction to dimethylhydrazine (UDMH), enabling ultrasensitive detection of UDMH (10-13 M). Importantly, ocSERS could not only monitor elevated levels of NDMA in ranitidine due to improper storage but also quantify NDMA in urine and blood after oral administration of NDMA-containing drugs, thereby preventing NDMA overexposure. Therefore, ocSERS represents the first click SERS sensor and may open up a new analytical field.
Collapse
Affiliation(s)
- Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing210093, China
| | - Xiaofeng Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| | - Chaowei Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou350108, Fujian, China
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Health Science center, Xi'an Jiaotong University, Xi'an710061, China
| | - Wenting Yang
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| | - Jielei He
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| | - Tianliang Fang
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| | - Weiwei Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| | - Haixiong Ge
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| | - Zhuo Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing210093, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing210093, China
| |
Collapse
|
11
|
Horne S, Vera MD, Nagavelli LR, Sayeed VA, Heckman L, Johnson D, Berger D, Yip YY, Krahn CL, Sizukusa LO, Rocha NFM, Bream RN, Ludwig J, Keire DA, Condran G. Regulatory Experiences with Root Causes and Risk Factors for Nitrosamine Impurities in Pharmaceuticals. J Pharm Sci 2023; 112:1166-1182. [PMID: 36599405 DOI: 10.1016/j.xphs.2022.12.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 01/02/2023]
Abstract
N-Nitrosamines (also referred to as nitrosamines) are a class of substances, many of which are highly potent mutagenic agents which have been classified as probable human carcinogens. Nitrosamine impurities have been a concern within the pharmaceutical industry and by regulatory authorities worldwide since June 2018, when regulators were informed of the presence of N-nitrosodimethylamine (NDMA) in the angiotensin-II receptor blocker (ARB) medicine, valsartan. Since that time, regulatory authorities have collaborated to share information and knowledge on issues related to nitrosamines with a goal of promoting convergence on technical issues and reducing and mitigating patient exposure to harmful nitrosamine impurities in human drug products. This paper shares current scientific information from a quality perspective on risk factors and potential root causes for nitrosamine impurities, as well as recommendations for risk mitigation and control strategies.
Collapse
Affiliation(s)
| | - Matthew D Vera
- US Food and Drug Administration (US FDA), Silver Spring, MD, 20993, USA
| | - Laxma R Nagavelli
- US Food and Drug Administration (US FDA), Silver Spring, MD, 20993, USA
| | - Vilayat A Sayeed
- US Food and Drug Administration (US FDA), Silver Spring, MD, 20993, USA
| | - Laurel Heckman
- US Food and Drug Administration (US FDA), Silver Spring, MD, 20993, USA
| | - Deborah Johnson
- US Food and Drug Administration (US FDA), Silver Spring, MD, 20993, USA
| | - Dan Berger
- US Food and Drug Administration (US FDA), Silver Spring, MD, 20993, USA
| | | | | | | | | | - Robert N Bream
- European Medicines Agency (EMA, EU), Amsterdam, the Netherlands
| | - Joachim Ludwig
- Bundesinstitut für Arzneimittel und Medizinprodukte (BfArM), Bonn, Germany
| | - David A Keire
- US Food and Drug Administration (US FDA), St Louis, MO, 63110, USA
| | | |
Collapse
|
12
|
Leigh Wood E, Tatke A, Viehmann A, Ashtiani M, Friedman RL, Kopcha M, Fisher AC. Dosage unit uniformity and dissolution testing of extended-release pharmaceutical products marketed in the U.S. Int J Pharm 2022; 625:122119. [PMID: 36029995 DOI: 10.1016/j.ijpharm.2022.122119] [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: 05/10/2022] [Revised: 08/03/2022] [Accepted: 08/12/2022] [Indexed: 10/15/2022]
Abstract
An international sampling study yielded 69 samples of extended-release prescription pharmaceuticals for legal sale in the U.S. Samples included 29 lots of innovator and 40 lots of generic solid oral extended-release drugs manufactured at 16 different facilities and containing 6 different active ingredients. Dosage unit uniformity and dissolution were tested for each lot. All samples met the relevant testing criteria for dosage unit uniformity and dissolution. There were no indications that manufacturer or region impacted a product's acceptability for use by patients. The variability of attributes was used to calculate a process performance index (Ppk) for each facility. Higher Ppk values suggest less variability relative to specification limits. Only two manufacturers fell below a 4-sigma manufacturing benchmark Ppk of 1.33 for dosage unit uniformity: a European manufacturer of a brand drug and an Asian manufacturer of a generic drug. Conversely, all but four manufacturers fell below a 4-sigma benchmark for the minimum Ppk across their product's dissolution timepoints: generic drug manufacturers in India (two), the U.S., and Canada. Compared to the immediate-release products of a previous study, Ppks were generally lower for extended-release products. A retrospective analysis found that manufacturers performing below median Ppks submitted more Field Alert Reports after the end of the sampling period.
Collapse
Affiliation(s)
- Erin Leigh Wood
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - Akshaya Tatke
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - Alex Viehmann
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - Melika Ashtiani
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - Richard L Friedman
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - Michael Kopcha
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - Adam C Fisher
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA.
| |
Collapse
|
13
|
Kamal AN, Dhar SI, Bock JM, Clarke JO, Lechien JR, Allen J, Belafsky PC, Blumin JH, Chan WW, Fass R, Fisichella PM, Marohn M, O'Rourke AK, Postma G, Savarino EV, Vaezi MF, Carroll TL, Akst LM. Best Practices in Treatment of Laryngopharyngeal Reflux Disease: A Multidisciplinary Modified Delphi Study. Dig Dis Sci 2022; 68:1125-1138. [PMID: 35995882 DOI: 10.1007/s10620-022-07672-9] [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] [Received: 05/10/2022] [Accepted: 08/12/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Laryngopharyngeal reflux (LPR) is a common otolaryngologic diagnosis. Treatment of presumed LPR remains challenging, and limited frameworks exist to guide treatment. METHODS Using RAND/University of California, Los Angeles (UCLA) Appropriateness Methods, a modified Delphi approach identified consensus statements to guide LPR treatment. Experts independently and blindly scored proposed statements on importance, scientific acceptability, usability, and feasibility in a four-round iterative process. Accepted measures reached scores with ≥ 80% agreement in the 7-9 range (on a 9-point Likert scale) across all four categories. RESULTS Fifteen experts rated 36 proposed initial statements. In round one, 10 (27.8%) statements were rated as valid. In round two, 8 statements were modified based on panel suggestions, and experts subsequently rated 5 of these statements as valid. Round three's discussion refined statements not yet accepted, and in round four, additional voting identified 2 additional statements as valid. In total, 17 (47.2%) best practice statements reached consensus, touching on topics as varied as role of empiric treatment, medication use, lifestyle modifications, and indications for laryngoscopy. CONCLUSION Using a well-tested methodology, best practice statements in the treatment of LPR were identified. The statements serve to guide physicians on LPR treatment considerations.
Collapse
Affiliation(s)
- Afrin N Kamal
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Redwood City, CA, USA.
| | - Shumon I Dhar
- Department of Otolaryngology, Head and Neck Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan M Bock
- Department of Otolaryngology & Communication Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John O Clarke
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Redwood City, CA, USA
| | - Jerome R Lechien
- Department of Otolaryngology Head Neck Surgery, Elsan Hospital, Paris, France
| | - Jacqueline Allen
- Department of Otolaryngology, Head and Neck Surgery, University of Auckland, Auckland, New Zealand
| | - Peter C Belafsky
- Department of Otolaryngology/Head and Neck Surgery, Davis School of Medicine, University of California, Sacramento, CA, USA
| | - Joel H Blumin
- Department of Otolaryngology & Communication Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Walter W Chan
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ronnie Fass
- Division of Gastroenterology, Department of Medicine, MetroHealth, Cleveland, OH, USA
| | | | - Michael Marohn
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ashli K O'Rourke
- Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Gregory Postma
- Department of Otolaryngology, Head and Neck Surgery, Augusta University, Augusta, GA, USA
| | - Edoardo V Savarino
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Michael F Vaezi
- Division of Gastroenterology, Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Thomas L Carroll
- Division of Otolaryngology and Harvard Medical School, Department of Otolaryngology-Head and Neck Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Lee M Akst
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Shin CM. Acid Suppressive Drugs. THE KOREAN JOURNAL OF HELICOBACTER AND UPPER GASTROINTESTINAL RESEARCH 2022. [DOI: 10.7704/kjhugr.2022.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Histamine H<sub>2</sub> receptor antagonists (H<sub>2</sub>RAs) suppress gastric acid production by blocking H<sub>2</sub> receptors in parietal cells. Studies have shown that proton pump inhibitors (PPIs) are superior to H<sub>2</sub>RAs as a treatment for acid-related disorders, such as peptic ulcer disease (PUD) and gastroesophageal reflux disease (GERD). PPIs reduce gastric acid production by irreversibly inhibiting the H<sup>+</sup>/K<sup>+</sup> ATPase pump, and they also increase gastric emptying. Although PPIs have differing pharmacokinetic properties, each PPI is effective in managing GERD and PUDs. However, PPIs have some limitations, including short plasma half-lives, breakthrough symptoms (especially at night), meal-associated dosing, and concerns associated with long-term PPI use. Potassium-competitive acid blockers (P-CABs) provide more rapid and profound suppression of intragastric acidity than PPIs. P-CABs are non-inferior to lansoprazole in healing erosive esophagitis and peptic ulcers, and may also be effective in improving symptoms in patients with non-erosive reflux disease. Acid suppressive drugs are the most commonly used drugs in clinical practice, and it is necessary to understand the pharmacological properties and adverse effects of each drug.
Collapse
|
15
|
Strauss DG, Florian J, Keire D. Risk of N-Nitrosodimethylamine (NMDA) Formation With Ranitidine. JAMA 2021; 326:2077. [PMID: 34812875 DOI: 10.1001/jama.2021.16261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- David G Strauss
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Jeffry Florian
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - David Keire
- Center for Drug Evaluation and Research, US Food and Drug Administration, St Louis, Missouri
| |
Collapse
|
16
|
White CM, Hernandez AV. Risk of N-Nitrosodimethylamine (NMDA) Formation With Ranitidine-Reply. JAMA 2021; 326:2077-2078. [PMID: 34812870 DOI: 10.1001/jama.2021.16264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- C Michael White
- Health Outcomes, Policy, and Evidence Synthesis (HOPES) Group, University of Connecticut School of Pharmacy, Storrs
| | - Adrian V Hernandez
- Health Outcomes, Policy, and Evidence Synthesis (HOPES) Group, University of Connecticut School of Pharmacy, Storrs
| |
Collapse
|
17
|
Adami HO, Trolle Andersen I, Heide-Jørgensen U, Chang ET, Nørgaard M, Toft Sørensen H. Ranitidine Use and Risk of Upper Gastrointestinal Cancers. Cancer Epidemiol Biomarkers Prev 2021; 30:2302-2308. [PMID: 34620629 DOI: 10.1158/1055-9965.epi-21-0831] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/01/2021] [Accepted: 09/30/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The discovery that ranitidine is contaminated with N-nitrosodimethylamine, a suspected human carcinogen, raises the hypothesis of a gastrointestinal carcinogenic effect; however, evidence remains inconclusive. METHODS We used the nationwide Danish Prescription Registry to identify a cohort of incident ranitidine users and two active comparator cohorts comprising users of other histamine-2 receptor blockers (H2RB) and users of proton pump inhibitors (PPI). All Danish adults with a first prescription of ranitidine, other H2RBs, or PPIs in 1996 through 2008 were followed virtually completely through 2018 for incidence of esophageal, stomach, liver, and pancreatic cancers. We used Cox regression with propensity-score weighting to calculate hazard ratios and 10-year cumulative risk with 95% confidence intervals. RESULTS We ascertained 276 newly diagnosed esophageal, 342 stomach, 133 hepatocellular, and 517 pancreatic cancers among ranitidine users during follow-up (median 14 years). In comparison with use of other H2RBs or PPIs, we found no consistent evidence of increased HRs or excess 10-year cumulative risk of any upper gastrointestinal cancer following ranitidine use. We observed no association after restriction to subjects with at least 5 or 10 prescriptions or those with 10 prescriptions and at least 10 years of follow-up. CONCLUSIONS Our large prospective study using high-quality prescription and cancer incidence data, with two active comparator groups, provides no compelling evidence that ranitidine increases the risk of upper gastrointestinal cancers. IMPACT Our results, which do not support any carcinogenic effect on esophagus, stomach, liver or pancreas, should be reassuring for millions of concerned past users of ranitidine.
Collapse
Affiliation(s)
- Hans-Olov Adami
- Clinical Effectiveness Group, Institute of Health and Society, University of Oslo, Oslo, Norway. .,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ina Trolle Andersen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Uffe Heide-Jørgensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ellen T Chang
- Center for Health Sciences, Exponent, Inc., Menlo Park, California.,Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mette Nørgaard
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henrik Toft Sørensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| |
Collapse
|