Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
In Vivo
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
In Vivo

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Visit iiar on Facebook
  • Follow us on Linkedin
Research ArticleClinical Studies
Open Access

Time-to-onset Analysis of Rhabdomyolysis due to Different Proton Pump Inhibitors Using a Pharmacovigilance Database

KATSUHIRO OHYAMA, MEGUMI IIDA, SHOTA AKIYAMA, HIROSHI YAMAZAKI and YUSUKE HORI
In Vivo May 2024, 38 (3) 1285-1291; DOI: https://doi.org/10.21873/invivo.13567
KATSUHIRO OHYAMA
1Center for Experiential Pharmacy Practice, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: ohyamakt@toyaku.ac.jp
MEGUMI IIDA
1Center for Experiential Pharmacy Practice, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SHOTA AKIYAMA
1Center for Experiential Pharmacy Practice, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HIROSHI YAMAZAKI
2Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Tokyo, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YUSUKE HORI
1Center for Experiential Pharmacy Practice, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Recent research has increasingly demonstrated an association between proton pump inhibitors (PPIs) and serious adverse events. This study aimed to evaluate the association between PPI and rhabdomyolysis (RM), examining its time-to-onset profiles using the Japanese Adverse Drug Event Report (JADER) database. Patients and Methods: Data spanning from April 2004 to March 2022 were used. The association between PPIs and RM was evaluated using the reporting odds ratio (ROR), adjusted for sex and age. Subsequent analyses were conducted after excluding cases involving concomitant use of statins or fibrates. Furthermore, the onset time of RM and Weibull distribution parameters were calculated to evaluate the expression profile of RM, and the outcomes were examined. Results: RM was associated with the use of esomeprazole, omeprazole, and rabeprazole, even in the absence of concomitant statin or fibrate use. The median time to RM onset varied among PPIs, ranging from 6.5 to 127 d. The Weibull distribution parameters indicated that the hazard types of nearly all orally administered PPIs were classified as early failure or close to random failure. Regarding outcomes, cases of death were reported for all PPIs except vonoprazan. Conclusion: The findings suggest the need for vigilant monitoring of RM during PPI administration, particularly in the early stages, considering the varying onset times.

Key Words:
  • Rhabdomyolysis
  • proton pump inhibitors
  • disproportionality analysis
  • adverse event profiles
  • Weibull distribution
  • Japanese Adverse Drug Event Report database

Drug-induced myopathies can arise through various mechanisms, including direct myotoxicity associated with substances like corticosteroids, alcohol, cocaine, colchicine, statins, and antimalarials. Additionally, immunologically induced inflammatory myopathy can occur with statins, penicillamine, interferon-alfa/interferon-beta, TNF-alpha blockers, immune checkpoint inhibitors, and monoclonal antibodies. Another mechanism involves indirect skeletal muscle tissue injury resulting from multifactorial etiologies, such as drug-induced comas leading to ischemic muscle compression or drug-induced hypokalemia (1, 2). The clinical manifestations of drug-induced myopathies can vary widely, ranging from asymptomatic or mild myalgias with or without muscle weakness to chronic myopathy with severe weakness and in rare cases, it may progress to more severe conditions like rhabdomyolysis (RM) (1, 3). RM is characterized by muscle necrosis and the release of cell degradation products and intracellular elements into the bloodstream and extracellular space (3, 4). The incidence of acute kidney injury, the most significant consequence of RM, varies from 13% to over 50%, depending on the cause, clinical context, and histological findings at the time of diagnosis (4).

Proton pump inhibitors (PPIs) are frequently prescribed medications for the management of gastric acid-related conditions, such as gastroesophageal reflux disease, Helicobacter pylori-induced gastric ulcers, duodenal ulcers, erosive esophagitis, and Zollinger–Ellison syndrome (5, 6). In addition to the common adverse events (AEs) listed in package inserts, recent studies have provided growing evidence of the association between PPIs and severe AEs, including kidney injury (7, 8), bone fractures (9), and Clostridium difficile-associated diarrhea (10).

In addition to the previously mentioned AEs, RM is also identified as a severe AE linked to the use of PPIs (11-13). Currently, mild myalgias are categorized as rare AEs for the overall PPI class, but the development of RM is not recognized as a known consequence of PPI usage (14). Notably, not all package inserts for each PPI available in Japan include RM as a listed AE, raising uncertainty about whether this AE is a class effect of PPIs. Moreover, there is limited information on the time to onset of RM in patients undergoing PPI treatment. Therefore, this study aimed to evaluate the association between PPIs and RM and the time-to-onset profiles of PPIs using the Japanese Adverse Drug Event Report (JADER) database.

Patients and Methods

Study data. The JADER data spanning from the first quarter of 2004 to the second quarter of 2022 were downloaded from the Pharmaceuticals and Medical Devices Agency website (15). The data encompassed four types of information: patient demographics (DEMO), drug treatments (DRUG), AEs (REAC), and medical history and primary disease information (HIST). The DEMO table contained basic patient details like reporting year, sex, and age, with age described in decades (e.g., 10s, 20s, etc.). For this study, reports with incomplete, unclear, or ambiguous age categories (e.g., adult, child, aged, and newborn) were excluded. Only reports with age stratification (<10s, 10s, 20s, 30s, 40s, 50s, 60, 70s, 80s, and ≥90s) were utilized. The DRUG table included details about the drug name, route of administration, start and end dates of administration, and its association with AEs. The REAC table provided information on AEs, including their names, onset dates, and outcomes. In this study, DEMO, DRUG, and REAC tables were used for analysis.

Drug of interest. The drugs of interest included esomeprazole, lansoprazole, omeprazole, rabeprazole, and vonoprazan. Drugs intended for H. pylori eradication, including combined drugs, were excluded. Within the DRUG table, each drug’s causality was categorized using codes indicating its association with adverse drug reactions, such as “suspected drug”, “concomitant drug”, or “interacting drug”. In this study, only “suspected drug” was used for the analysis.

Definition of RM. RM was defined based on the preferred term “rhabdomyolysis” (PT 10039020) in the Medical Dictionary for Regulatory Activities (MedDRA/J ver. 25.1).

Disproportionality analysis. The association between PPIs and RM was evaluated using the reporting odds ratio (ROR) (16-18). Signal scores were determined through the case/non-case method (19, 20).

To account for covariates and enable adjustment via logistic regression analysis (21-23), the adjusted ROR (aROR) was calculated following established methodologies (24, 25). Only reports containing complete information on sex and age were considered for aROR calculation. Additional analyses were conducted, excluding reports involving concomitant use of statins (atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin) or fibrates (clofibrate, fenofibrate, bezafibrate, and pemafibrate) for further assessment.

An association was defined when the lower limit of the 95% confidence interval (CI) of the aROR was >1, and the p-value was <0.05.

Time to onset of RM. The calculation of the onset time of RM involved determining the number of days from the initiation of the administration of the drug of interest to the onset of RM, using time information from the DRUG and REAC tables. Therefore, only reports containing available time-to-onset data were analyzed. The median days (interquartile range: IQR; no. d) for the onset of RM were calculated based on the administration routes.

Outcomes after RM onset. There are six outcome descriptors in the REAC table of the JADER: “recovery”, “remission”, “with sequelae”, “not recovered”, “death”, and “unclear”.

Weibull distribution. Furthermore, the Weibull parameters from the Weibull distribution were used to evaluate the AE profile (26-28). The Weibull distribution is expressed using the scale parameter α, representing the scale of the distribution function, and the shape parameter β, indicating the change in hazard without a reference population over time. The shape parameter β value categorizes failure into three groups: a β value with a 95%CI <1 indicates an initial increase in hazard followed by a decrease (early failure type); a β value close to or equal to 1, with a 95%CI of 1, indicates a constant hazard throughout the exposure period (random failure type); and a β value with a 95% CI >1 signifies an increasing hazard over time (wear-out failure type).

All statistical analyses and data visualization were conducted using JMP Pro ver. 13.2.1 (SAS Institute, Cary, NC, USA). Significance was considered for p-values <0.05, and an aROR with the lower limit of the 95%CI >1 was deemed significant.

Results

Data analyzed. In total, 777,555 reports were extracted from the JADER database. Following the exclusion of records with missing, unknown, or inadequate data, only reports involving suspected drugs were considered. Finally, 618,338 reports were analyzed, including 5,962 RM reports.

Association between PPI use and RM. Table I shows the analysis of the association between PPIs and RM. Esomeprazole, omeprazole, and rabeprazole were associated with RM (Table I). Even after excluding users of concomitant statins or fibrates, the aROR values exhibited no significant change, and the association between each drug and RM remained consistent (Table I).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Association of rhabdomyolysis with proton pump inhibitor treatment with and without concomitant use of statins or fibrates.

Time to onset of RM. Figure 1 displays box plots illustrating the onset times of RM. The median day (IQR) for RM onset for the intravenous administration route (only omeprazole was calculable) was 2 [1-6] d. Conversely, for the oral administration route, the smallest value for RM onset was 6.5 [2-25] d for esomeprazole, while the largest value was 127 [17.5-196] d for omeprazole (Figure 1).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Box plot illustrating the onset time of rhabdomyolysis after the administration of proton pump inhibitors. Lines within the boxes represent the median, the boxes depict the interquartile range, and the whiskers indicate the minimum and maximum values.

Weibull distribution analysis. Table II shows the results of the Weibull distribution analysis for PPIs. For the oral administration route, esomeprazole, lansoprazole, and rabeprazole were indicated to have an early failure-type profile, whereas others exhibited a random failure-type profile (Table II).

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table II.

Weibull parameters for rhabdomyolysis as an adverse event of treatment with proton pump inhibitor.

Outcomes after RM onset. Table III displays the number and percentage of the six outcomes following the onset RM. Omeprazole was the most frequently reported drug, observed in both oral and intravenous routes. Across oral route medications, the reported numbers were nearly equal for all PPIs, with recovery and remission accounting for more than 80% of the cases. It is important to note that deaths were reported for all drugs except vonoprazan, albeit in small numbers.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table III.

The number and percentage of rhabdomyolysis outcomes by route of proton pump inhibitor administration.

Discussion

Our findings suggest an association of certain PPIs, such as esomeprazole, omeprazole, and rabeprazole, with RM even when concomitant use of statins or fibrates is excluded. The median onset day for RM via intravenous administration was 2 [1-6] d for omeprazole, whereas for the oral administration route, the smallest median ranged from 6.5 [2-25] d for esomeprazole to 127 [17.5-196] d for omeprazole, the highest median observed. The Weibull distribution analysis revealed early failure-type profiles for esomeprazole, lansoprazole, and rabeprazole, while others exhibited random failure-type profiles. Regarding outcomes post-RM onset, almost all cases were favorable; however, although the numbers were small, deaths were reported for all drugs, except vonoprazan.

While the precise mechanism of RM following PPI use remains uncertain, various hypotheses have been proposed, including the potential development of electrolyte imbalances or autoimmune antibodies as a result of long-term PPI use (29, 30). In particular, two mechanisms have been under consideration. First, it has been suggested that PPIs bind to H+/K+-ATPase in gastric parietal cells. As H+/K+-ATPase regulates intracellular pH, this interaction could enhance susceptibility to cellular degradation, thereby increasing the risk of RM (31). Another hypothesis revolves around the potential of PPIs to induce autoimmune disorders in patients with familial myopathy, subsequently leading to autoimmune muscle diseases like polymyositis or myasthenia gravis (32, 33).

In Japan, RM is listed in the package inserts for esomeprazole, omeprazole, and rabeprazole, but not for lansoprazole or vonoprazan. Therefore, our findings indicate that each package insert appropriately reflects the observed association between the respective drug and RM. In contrast, a disproportionality analysis using the FDA Adverse Event Reporting System (FAERS), the largest spontaneous reporting database, demonstrated an association between RM and all PPIs, including pantoprazole (34). However, this study using the JADER differs from previous reports concerning lansoprazole and vonoprazan. Further studies are needed to clarify whether the occurrence of RM following PPI use represents a class effect of PPIs, including understanding its underlying mechanism.

Given that statins and fibrates are commonly implicated in RM development (1, 35), we explored the impact of concomitant statin use in patients receiving PPI. Some statins serve as substrates for CYP3A4, the enzyme responsible for PPI sulfoxidation (36, 37). Additionally, CYP2C19, exhibiting genetic polymorphism in approximately 20% of East Asians, is accountable for the primary metabolic pathway of PPIs, involving 5-hydroxylation, except for vonoprazan. Therefore, the concomitant use of both drugs is anticipated to raise the likelihood of RM. Nevertheless, the effect of concomitant statin usage on the association between PPIs and this AE appeared to be limited. Similar findings have been observed in analyses of other spontaneous AE databases, including the FAERS and the Italian National Network of Pharmacovigilance database (14, 34). This is likely attributed to the distinct mechanisms underlying PPI-induced RM compared to that caused by statins, coupled with the lower incidence of PPI-induced RM in comparison to that induced by statins.

In this study, the time-to-onset of RM varied across PPIs. The largest median value with IQR was 127 [17.5-196] d for omeprazole, while the smallest value was 6.5 [2-25] d for oral esomeprazole (Figure 1). This variability could be attributed to differences in the involvement of CYP2C19 in the metabolism of each drug. Since esomeprazole consists only of the S (-) form, the contribution of CYP2C19 is limited, with CYP3A4 playing a predominant role in metabolism. On the contrary, omeprazole, a racemic form, may have a more complex metabolism due to the presence of the R (-) form and the varying contribution of CYP2C19 and CYP3A4 among individuals, especially in East Asians.

There is limited available information on the time-to-onset profile of RM related to PPIs. In this study, the analyses of median onset time and Weibull distribution for the intravenous administration route, although focused on a single drug, revealed a median of 2 [1-6] d characterized by a random failure type. For the oral administration route, the onset time varied from 6.5 [2-25] d to 127 [17.5-196] d, displaying early failure/random failure types (tending toward early failure) (Figure 1). This suggests that healthcare providers should closely monitor patients throughout the course of intravenous administration, particularly in the early stages following oral administration initiation, for the occurrence of RM.

Regarding the outcomes following the oral administration of RM, “Recovery” and “Remission” constituted more than 80%-90% for each orally administered drug, while “Death” ranged from 3.4% to 9.7% for each drug, excluding vonoprazan (Table III). This aligns with general reports indicating that approximately 5% of RM patients experience worse outcomes (38), consistent with previous findings. In contrast, though the results primarily involving omeprazole, the occurrence of “Death” was approximately 17% for patients treated intravenously (Table III). The primary contributing factor to this difference may be the patient’s condition. As highlighted earlier, RM following intravenous PPI administration unfolds within a confined and brief timeframe, underscoring the importance of vigilant monitoring during this period.

While using a spontaneous reporting system has several advantages in detecting potential drug–AE associations, including access to information regarding AE onset times, its inherent characteristics introduce potential limitations that might impact the interpretation of our findings. First, it remains uncertain whether the reported AEs can be definitively attributed to the administration of a specific medication. Second, RM can be influenced by various concomitant drugs, other than statins and fibrates. Although the PPIs examined in this study were designated as “suspected drugs”, the impact of concomitant drugs known to induce myopathy, such as corticosteroids, colchicine, penicillamine, and other anticancer drugs, was not considered (1, 2). Third, not every AE associated with a drug may be reported (i.e., reporting bias). Finally, the exclusion of some data was necessary due to the missing information that rendered the calculation of onset times impossible.

Conclusion

Our results suggest that the time-to-onset profiles of RM due to PPI differ depending on the drug and administration route. Considering the potential fatality associated with RM during PPI administration, healthcare providers should exercise careful monitoring, especially during intravenous administration and the early stages of oral administration. This vigilance is crucial considering the different onset times associated with different PPIs.

Footnotes

  • Authors’ Contributions

    K. Ohyama and Y. Hori conceived of and designed the study. K. Ohyama, M. Iida, and S. Akiyama analyzed the data. K. Ohyama and H. Yamazaki drafted the manuscript. All Authors critically reviewed and approved the final version of the manuscript.

  • Funding

    This research received no specific grants from any funding agency in the public, commercial, or not-for-profit sectors.

  • Conflicts of Interest

    The Authors declare that they have no conflicts of interest in relation to this study.

  • Received January 12, 2024.
  • Revision received February 14, 2024.
  • Accepted February 15, 2024.
  • Copyright © 2024, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Ostrowski P,
    2. Bonczar M,
    3. Avram AE,
    4. Lippi G,
    5. Henry BM
    : Safety monitoring of drug-induced muscle injury and rhabdomyolysis: a biomarker-guided approach for clinical practice and drug trials. Clin Chem Lab Med 61(10): 1688-1699, 2023. DOI: 10.1515/cclm-2023-0313
    OpenUrlCrossRef
  2. ↵
    1. Prendergast BD,
    2. George CF
    : Drug-induced rhabdomyolysis—mechanisms and management. Postgrad Med J 69(811): 333-336, 1993. DOI: 10.1136/pgmj.69.811.333
    OpenUrlFREE Full Text
  3. ↵
    1. Alfirevic A,
    2. Neely D,
    3. Armitage J,
    4. Chinoy H,
    5. Cooper RG,
    6. Laaksonen R,
    7. Carr DF,
    8. Bloch KM,
    9. Fahy J,
    10. Hanson A,
    11. Yue QY,
    12. Wadelius M,
    13. Maitland-van Der Zee AH,
    14. Voora D,
    15. Psaty BM,
    16. Palmer CN,
    17. Pirmohamed M
    : Phenotype standardization for statin-induced myotoxicity. Clin Pharmacol Ther 96(4): 470-476, 2014. DOI: 10.1038/clpt.2014.121
    OpenUrlCrossRefPubMed
  4. ↵
    1. Cervellin G,
    2. Comelli I,
    3. Benatti M,
    4. Sanchis-Gomar F,
    5. Bassi A,
    6. Lippi G
    : Non-traumatic rhabdomyolysis: Background, laboratory features, and acute clinical management. Clin Biochem 50(12): 656-662, 2017. DOI: 10.1016/j.clinbiochem.2017.02.016
    OpenUrlCrossRef
  5. ↵
    1. Song H,
    2. Zhu J,
    3. Lu D
    : Long-term proton pump inhibitor (PPI) use and the development of gastric pre-malignant lesions. Cochrane Database Syst Rev 2014(12): CD010623, 2014. DOI: 10.1002/14651858.CD010623.pub2
    OpenUrlCrossRef
  6. ↵
    1. Makunts T,
    2. Alpatty S,
    3. Lee KC,
    4. Atayee RS,
    5. Abagyan R
    : Proton-pump inhibitor use is associated with a broad spectrum of neurological adverse events including impaired hearing, vision, and memory. Sci Rep 9(1): 17280, 2019. DOI: 10.1038/s41598-019-53622-3
    OpenUrlCrossRefPubMed
  7. ↵
    1. Lazarus B,
    2. Chen Y,
    3. Wilson FP,
    4. Sang Y,
    5. Chang AR,
    6. Coresh J,
    7. Grams ME
    : Proton Pump Inhibitor Use and the Risk of Chronic Kidney Disease. JAMA Intern Med 176(2): 238-246, 2016. DOI: 10.1001/jamainternmed.2015.7193
    OpenUrlCrossRefPubMed
  8. ↵
    1. Xie Y,
    2. Bowe B,
    3. Li T,
    4. Xian H,
    5. Balasubramanian S,
    6. Al-Aly Z
    : Proton Pump Inhibitors and Risk of Incident CKD and Progression to ESRD. J Am Soc Nephrol 27(10): 3153-3163, 2016. DOI: 10.1681/ASN.2015121377
    OpenUrlAbstract/FREE Full Text
  9. ↵
    1. Wilson C
    : Bone: proton-pump inhibitors and fractures. Nat Rev Endocrinol 8(11): 625-625, 2012. DOI: 10.1038/nrendo.2012.170
    OpenUrlCrossRefPubMed
  10. ↵
    1. Janarthanan S,
    2. Ditah I,
    3. Adler DG,
    4. Ehrinpreis MN
    : Clostridium difficile-associated diarrhea and proton pump inhibitor therapy: a meta-analysis. Am J Gastroenterol 107(7): 1001-1010, 2012. DOI: 10.1038/ajg.2012.179
    OpenUrlCrossRefPubMed
  11. ↵
    1. Nozaki M,
    2. Suzuki T,
    3. Hirano M
    : Rhabdomyolysis associated with omeprazole. J Gastroenterol 39(1): 86-86, 2004. DOI: 10.1007/s00535-003-1231-7
    OpenUrlCrossRefPubMed
    1. Tanaka K,
    2. Nakada TA,
    3. Abe R,
    4. Itoga S,
    5. Nomura F,
    6. Oda S
    : Omeprazole-associated rhabdomyolysis. Crit Care 18(4): 462, 2014. DOI: 10.1186/s13054-014-0462-8
    OpenUrlCrossRefPubMed
  12. ↵
    1. Tröger U,
    2. Reiche I,
    3. Jepsen MS,
    4. Huth C,
    5. Bode-Böger SM
    : Esomeprazole-induced rhabdomyolysis in a patient with heart failure. Intensive Care Med 36(7): 1278-1279, 2010. DOI: 10.1007/s00134-010-1854-0
    OpenUrlCrossRefPubMed
  13. ↵
    1. Capogrosso Sansone A,
    2. Convertino I,
    3. Galiulo MT,
    4. Salvadori S,
    5. Pieroni S,
    6. Knezevic T,
    7. Mantarro S,
    8. Marino A,
    9. Hauben M,
    10. Blandizzi C,
    11. Tuccori M
    : Muscular adverse drug reactions associated with proton pump inhibitors: a disproportionality analysis using the Italian National Network of Pharmacovigilance Database. Drug Saf 40(10): 895-909, 2017. DOI: 10.1007/s40264-017-0564-8
    OpenUrlCrossRef
  14. ↵
    Terms of use for Japanese Adverse Drug Event Reporting system: The Pharmaceuticals and Medical Devices Agency website. Available at: https://www.pmda.go.jp/safety/info-services/drugs/adr-info/suspected-adr/0003.html [Last accessed on July 11, 2022]
  15. ↵
    1. Kimura K,
    2. Kikegawa M,
    3. Kan Y,
    4. Uesawa Y
    : Identifying Crude Drugs in Kampo Medicines Associated with Drug-Induced Liver Injury Using the Japanese Adverse Drug Event Report Database: A Comprehensive Survey. Pharmaceuticals (Basel) 16(5): , 2023. DOI: 10.3390/ph16050678
    OpenUrlCrossRef
    1. Hosomi K,
    2. Fujimoto M,
    3. Ushio K,
    4. Mao L,
    5. Kato J,
    6. Takada M
    : An integrative approach using real-world data to identify alternative therapeutic uses of existing drugs. PLoS One 13(10): e0204648, 2018. DOI: 10.1371/journal.pone.0204648
    OpenUrlCrossRef
  16. ↵
    1. Ohyama K,
    2. Shindo J,
    3. Takahashi T,
    4. Takeuchi H,
    5. Hori Y
    : Pharmacovigilance study of the association between dipeptidyl peptidase-4 inhibitors and angioedema using the FDA Adverse Event Reporting System (FAERS). Sci Rep 12(1): 13122, 2022. DOI: 10.1038/s41598-022-17366-x
    OpenUrlCrossRef
  17. ↵
    1. Sakaeda T,
    2. Tamon A,
    3. Kadoyama K,
    4. Okuno Y
    : Data mining of the public version of the FDA Adverse Event Reporting System. Int J Med Sci 10(7): 796-803, 2013. DOI: 10.7150/ijms.6048
    OpenUrlCrossRefPubMed
  18. ↵
    1. Almenoff JS,
    2. Pattishall EN,
    3. Gibbs TG,
    4. DuMouchel W,
    5. Evans SJ,
    6. Yuen N
    : Novel statistical tools for monitoring the safety of marketed drugs. Clin Pharmacol Ther 82(2): 157-166, 2007. DOI: 10.1038/sj.clpt.6100258
    OpenUrlCrossRefPubMed
  19. ↵
    1. Van Puijenbroek EP,
    2. Egberts AC,
    3. Meyboom RH,
    4. Leufkens HG
    : Signalling possible drug-drug interactions in a spontaneous reporting system: delay of withdrawal bleeding during concomitant use of oral contraceptives and itraconazole. Br J Clin Pharmacol 47(6): 689-693, 1999. DOI: 10.1046/j.1365-2125.1999.00957.x
    OpenUrlCrossRefPubMed
    1. van Puijenbroek EP,
    2. Egberts AC,
    3. Heerdink ER,
    4. Leufkens HG
    : Detecting drug–drug interactions using a database for spontaneous adverse drug reactions: an example with diuretics and non-steroidal anti-inflammatory drugs. Eur J Clin Pharmacol 56(9-10): 733-738, 2000. DOI: 10.1007/s002280000215
    OpenUrlCrossRefPubMed
  20. ↵
    1. Ohyama K,
    2. Tanaka H,
    3. Hori Y
    : Effect of Concomitant Drug Use on the Onset and Exacerbation of Diabetes Mellitus in Everolimus-Treated Cancer. J Pharm Pharm Sci 25: 245-252, 2022. DOI: 10.18433/jpps32908
    OpenUrlCrossRefPubMed
  21. ↵
    1. Kawada K,
    2. Ishida T,
    3. Jobu K,
    4. Ohta T,
    5. Fukuda H,
    6. Morisawa S,
    7. Kawazoe T,
    8. Tamura N,
    9. Miyamura M
    : Association of aggression and antiepileptic drugs: Analysis using the Japanese Adverse Drug Event Report (JADER) database. Biol Pharm Bull 45(6): 720-723, 2022. DOI: 10.1248/bpb.b21-00954
    OpenUrlCrossRef
  22. ↵
    1. Ohyama K,
    2. Akiyama S,
    3. Iida M,
    4. Hori Y
    : Association of Torsade de Pointes and QT prolongation with antifungal triazoles: Analysis using a pharmacovigilance database. In Vivo 37(6): 2719-2725, 2023. DOI: 10.21873/invivo.13382
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Sauzet O,
    2. Carvajal A,
    3. Escudero A,
    4. Molokhia M,
    5. Cornelius VR
    : Illustration of the weibull shape parameter signal detection tool using electronic healthcare record data. Drug Saf 36(10): 995-1006, 2013. DOI: 10.1007/s40264-013-0061-7
    OpenUrlCrossRefPubMed
    1. Kawahara Y,
    2. Murata S,
    3. Shimizu T,
    4. Uesawa Y,
    5. Uchida M
    : Assessment of time-to-onset and outcome of lung adverse events with pomalidomide from a pharmacovigilance study. In Vivo 37(2): 955-961, 2023. DOI: 10.21873/invivo.13168
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Kan Y,
    2. Nagai J,
    3. Uesawa Y
    : Evaluation of antibiotic-induced taste and smell disorders using the FDA adverse event reporting system database. Sci Rep 11(1): 9625, 2021. DOI: 10.1038/s41598-021-88958-2
    OpenUrlCrossRef
  25. ↵
    1. Bebarta VS,
    2. King JA,
    3. McDonough M
    : Proton pump inhibitor–induced rhabdomyolysis and hyponatremic delirium. Am J Emerg Med 26(4): 519.e1-519.e2, 2008. DOI: 10.1016/j.ajem.2007.08.026
    OpenUrlCrossRefPubMed
  26. ↵
    1. Grattagliano I,
    2. Portincasa P,
    3. Mastronardi M,
    4. Palmieri VO,
    5. Palasciano G
    : Esomeprazole-induced central fever with severe myalgia. Ann Pharmacother 39(4): 757-760, 2005. DOI: 10.1345/aph.1E377
    OpenUrlCrossRefPubMed
  27. ↵
    1. Duncan SJ,
    2. Howden CW
    : Proton pump inhibitors and risk of rhabdomyolysis. Drug Saf 40(1): 61-64, 2017. DOI: 10.1007/s40264-016-0473-2
    OpenUrlCrossRef
  28. ↵
    1. Sivakumar K,
    2. Dalakas M
    : Autoimmune syndrome induced by omeprazole. Lancet 344(8922): 619-620, 1994. DOI: 10.1016/s0140-6736(94)92008-7
    OpenUrlCrossRefPubMed
  29. ↵
    1. Clark DWJ,
    2. Strandell J
    : Myopathy including polymyositis: a likely class adverse effect of proton pump inhibitors? Eur J Clin Pharmacol 62(6): 473-479, 2006. DOI: 10.1007/s00228-006-0131-1
    OpenUrlCrossRefPubMed
  30. ↵
    1. Mitsuboshi S,
    2. Hamano H,
    3. Kuniki Y,
    4. Niimura T,
    5. Chuma M,
    6. Ushio S,
    7. Lin T,
    8. Matsumoto J,
    9. Takeda T,
    10. Kajizono M,
    11. Zamami Y,
    12. Ishizawa K
    : Proton pump inhibitors and rhabdomyolysis: analysis of two different cross-sectional databases. Ann Pharmacother 57(11): 1255-1263, 2023. DOI: 10.1177/10600280231156270
    OpenUrlCrossRef
  31. ↵
    1. Adachi K,
    2. Ohyama K,
    3. Tanaka Y,
    4. Sato T,
    5. Murayama N,
    6. Shimizu M,
    7. Saito Y,
    8. Yamazaki H
    : High hepatic and plasma exposures of atorvastatin in subjects harboring impaired cytochrome P450 3A4*16 modeled after virtual administrations and possibly associated with statin intolerance found in the Japanese adverse drug event report database. Drug Metab Pharmacokinet 49: 100486, 2023. DOI: 10.1016/j.dmpk.2022.100486
    OpenUrlCrossRef
  32. ↵
    1. Uno Y,
    2. Uehara S,
    3. Ushirozako G,
    4. Murayama N,
    5. Suemizu H,
    6. Yamazaki H
    : Cytochrome P450 1A2 and 2C enzymes autoinduced by omeprazole in dog hepatocytes and human HepaRG and HepaSH cells are involved in omeprazole 5-hydroxylation and sulfoxidation. Xenobiotica 53(6-7): 465-473, 2023. DOI:10.1080/00498254.2023.2266840
    OpenUrlCrossRef
  33. ↵
    1. Yamazaki H,
    2. Inoue K,
    3. Shaw PM.,
    4. Checovich WJ,
    5. Guengerich FP,
    6. Shimada T
    : Different contributions of cytochrome P450 2C19 and 3A4 in the oxidation of omeprazole by human liver microsomes: effects of contents of these two forms in individual human samples. J Pharmacol Exp Ther 283(2): 434-442, 1997.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    1. Boutaud O,
    2. Roberts LJ 2nd.
    : Mechanism-based therapeutic approaches to rhabdomyolysis-induced renal failure. Free Radic Biol Med 51(5): 1062-1067, 2011. DOI: 10.1016/j.freeradbiomed.2010.10.704
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

In Vivo: 38 (3)
In Vivo
Vol. 38, Issue 3
May-June 2024
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on In Vivo.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Time-to-onset Analysis of Rhabdomyolysis due to Different Proton Pump Inhibitors Using a Pharmacovigilance Database
(Your Name) has sent you a message from In Vivo
(Your Name) thought you would like to see the In Vivo web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
11 + 4 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Time-to-onset Analysis of Rhabdomyolysis due to Different Proton Pump Inhibitors Using a Pharmacovigilance Database
KATSUHIRO OHYAMA, MEGUMI IIDA, SHOTA AKIYAMA, HIROSHI YAMAZAKI, YUSUKE HORI
In Vivo May 2024, 38 (3) 1285-1291; DOI: 10.21873/invivo.13567

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Time-to-onset Analysis of Rhabdomyolysis due to Different Proton Pump Inhibitors Using a Pharmacovigilance Database
KATSUHIRO OHYAMA, MEGUMI IIDA, SHOTA AKIYAMA, HIROSHI YAMAZAKI, YUSUKE HORI
In Vivo May 2024, 38 (3) 1285-1291; DOI: 10.21873/invivo.13567
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Patients and Methods
    • Results
    • Discussion
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Anamorelin and Conduction Defects: A Literature Review and Analysis of the Japanese Pharmacovigilance Database
  • Google Scholar

More in this TOC Section

  • Surgical Treatment and Prognosis of Soft Tissue Sarcoma in Patients Aged 85 Years and Older
  • Neutrophil-to-Lymphocyte Ratio as a Biomarker for Postoperative Complications in Crohn’s Disease
  • IgA Nephropathy Associated With Infliximab Treatment in Patients With Crohn’s Disease: Study of IgA1 and IgA2 Expression in Glomeruli
Show more Clinical Studies

Similar Articles

Keywords

  • Rhabdomyolysis
  • proton pump inhibitors
  • disproportionality analysis
  • adverse event profiles
  • Weibull distribution
  • Japanese Adverse Drug Event Report database
In Vivo

© 2025 In Vivo

Powered by HighWire