Abstract
Extensive media coverage of the potential health benefits of vitamin D supplementation has translated into substantial increases in supplement sales over recent years. Yet, the potential for drug-vitamin D interactions is rarely considered. This systematic review of the literature was conducted to evaluate the extent to which drugs affect vitamin D status or supplementation alters drug effectiveness or toxicity in humans. Electronic databases were used to identify eligible peer-reviewed studies published through September 1, 2010. Study characteristics and findings were abstracted, and quality was assessed for each study. A total of 109 unique reports met the inclusion criteria. The majority of eligible studies were classified as Class C (non-randomized trials, case-control studies, or time series) or D (cross-sectional, trend, case report/series, or before-and-after studies). Only two Class C and three Class D studies were of positive quality. Insufficient evidence was available to determine whether lipase inhibitors, antimicrobial agents, antiepileptic drugs, highly active antiretroviral agents or H2 receptor antagonists alter serum 25(OH)D concentrations. Atorvastatin appears to increase 25(OH)D concentrations, while concurrent vitamin D supplementation decreases concentrations of atorvastatin. Use of thiazide diuretics in combination with calcium and vitamin D supplements may cause hypercalcemia in the elderly, or those with compromised renal function or hyperparathyroidism. Larger studies with stronger study designs are needed to clarify potential drug-vitamin D interactions, especially for drugs metabolized by cytochrome P450 3A4 (CYP3A4). Health care providers should be aware of the potential for drug-vitamin D interactions.
Keywords: vitamin D, drug-nutrient interactions
Introduction
Vitamin D, a steroid hormone precursor, is well known for its role in maintaining calcium homeostasis and normal bone structure. Recent evidence suggests that the vitamin may also play a role in a variety of other physiologic processes such as modulation of inflammatory pathways 1 and susceptibility to diabetes 2, cancer 3, and infectious 4 and cardiovascular 5 diseases. Extensive media coverage of the potential health benefits of vitamin D has translated into vitamin D supplement sales in the United States (US) increasing from $75 million in 2006 to $550 million in 2010 6. Supplemental vitamin D is available in doses that can be considered pharmacologic (≥400 IU) compared to the usual US dietary intake (approximately 160–200 IU/day 7), and thus may interact with several types of prescription medications 8, potentially altering drug effectiveness or toxicity. Conversely, certain drugs may alter vitamin D metabolism and status.
Supplemental vitamin D is available in two forms, cholecalciferol (vitamin D3) and ergocalciferol (vitamin D2). Vitamin D3 is produced endogenously in the skin upon exposure to ultraviolet (UV) radiation, and is found in fortified foods and foods of animal origin such as fish, eggs, and liver. Vitamin D2 is only available exogenously, primarily through consumption of plant foods, fortified foods and dietary supplements. The liver is the primary site for the initial hydroxylation reaction that converts both vitamin D2 and D3 to the main circulating form of vitamin D, 25-hydroxycholecalciferol (25(OH)D). This conversion occurs via hepatic 25-hydoxylases, which include the cytochrome P450 (CYP) enzymes 2R1, 3A4, and 27A1. The active steroid hormone form of vitamin D is 1,25-dihydroxycholecalciferol (1,25(OH)2D), which is formed from 25(OH)D at both the local tissue level and in the kidney by an additional hydroxylation of 25(OH)D via 1α-hydroxylase (CYP27B1) 9. Catabolism of vitamin D metabolites occurs via 24-hydroxylase (CYP24A1). Vitamin D metabolism is depicted in Figure 1.
Figure 1. Vitamin D metabolism.
Ovals denote metabolic enzymes, rectangles denote substrates.
As a steroid hormone, 1,25(OH)2D is involved in intracellular signaling through both rapid responses (initiation of membrane-associated signal transduction) and genomic responses (initiation/inhibition of transcription for genes containing a vitamin D response element) 10. In the slower genomic responses, binding of 1,25(OH)2D to the vitamin D receptor in the cytoplasm forms a heterodimer with the retinoid X receptor (RXR), which is then translocated into the nucleus where it binds to vitamin D receptor elements (VDRE) in the promoter region of certain genes and either activates or inhibits gene transcription (Figure 2). Gene expression profiling has shown that 1,25(OH)2D enhances transcription of several phase 1 11 and phase 2 12 biotransformation enzymes, as well as p-glycoprotein (also known as multidrug resistant protein 1) 13, enzymes which are involved in drug bioavailability and metabolism.
Figure 2. Vitamin D intracellular signaling pathways.
As a steroid hormone, 1,25(OH)2D is involved in intracellular signaling through both rapid responses (initiation of membrane-associated signal transduction as a result of 1,25(OH)2D binding to membrane-bound vitamin D receptors (mVDR)) and genomic responses (initiation/inhibition of transcription for genes containing a vitamin D response element (VDRE)). In the slower genomic responses, vitamin D metabolites can enter the cell either as 25(OH)D (through carrier-mediated endocytosis with megalin or cubilin as the primary carriers, and subsequent intracellular conversion to 1,25(OH)2D), or directly as the active 1,25(OH)2D. Binding of 1,25(OH)2D to the vitamin D receptor (VDR) in the cytoplasm forms a heterodimer with the retinoid X receptor (RXR), which is then translocated into the nucleus where it binds to VDREs in the promoter region of certain genes and either activates or inhibits gene transcription in complex with RNA polymerase (RNA Pol).
The metabolically active 1,25(OH)2D form is tightly regulated at the tissue level, and is present in circulation only in picomolar quantities, thus 25(OH)D is considered the more clinically relevant metabolite for assessing overall vitamin D status. Although the Dietary Reference Intakes for Calcium and Vitamin D report issued in 2011 by the Institute of Medicine proposes ≥20 ng/mL as the definition of sufficiency based solely on requirements to prevent osteoporosis 14, it has been hypothesized that serum 25(OH)D concentrations of ≥30–32 ng/mL (75–80 nmol/L) are optimal in healthy populations 15–17.
Lower 25(OH)D levels are commonly reported in obese individuals compared to normal weight subjects. These findings have been attributed to sequestration of the fat-soluble vitamin D in adipose tissue, the major storage site for vitamin D 18. At latitudes >40° (Minneapolis = 45° N), UV intensity is not strong enough to stimulate cholecalciferol synthesis in the skin during the winter months 19. Several studies show that 25(OH)D concentrations are higher in men than women, although the reasons for these differences are not known 20. In addition to low dietary/supplemental vitamin D intake and low UV exposure, other factors associated with suboptimal 25(OH)D levels include advanced age and darker skin pigmentation 20, 21.
The 25-hydroxylase CYP3A4, which converts ergo- and cholecalciferol to 25(OH)D, is also a phase I biotransformation enzyme for many drugs 22. In vitro studies indicate that as many of half of all therapeutic drugs are metabolized by CYP3A4, while other drugs may inhibit or induce CYP3A4 activity (Table 1) 23. CYP3A4 is active in the mucosal enterocytes in the intestines as well as hepatocytes 24, 25, therefore interactions between orally administered drugs and dietary/supplemental vitamin D intake may be more significant than for intravenously administered drugs or vitamin D synthesized as a result of UV exposure. The CYP3A4 gene also contains a vitamin D response element, and CYP3A4 expression is up-regulated in the presence of 1,25(OH)2D 26, 27. Thus, vitamin D may alter metabolism of drugs requiring CYP3A4 activation 13.
Table 1.
Examples of drugs that are activated by, inhibit or induce CYP3A4
Substrate for CYP3A4 23, 173 | Inhibits CYP3A4 23, 173, 174 | Induces CYP3A4 23, 166, 167, 173 |
---|---|---|
Analgesics:
|
Antidiabetics:
|
Anticonvulsants
Antimicrobial agents:
Diuretics:
|
Indicates drugs that are reversible inhibitors of CYP3A4
indicates drugs that are able to induce their own metabolism
Other potential biologic mechanisms for drug-vitamin D interactions include: 1) altered absorption of the fat soluble vitamin D when taken concurrently with drugs that inhibit absorption or enhance elimination of dietary fat, and 2) exacerbation of risk of hypercalcemia when taken with calcium-sparing medications.
The purpose of this systematic review is to determine the extent to which drugs affect vitamin D status (by altering absorption, metabolism, or excretion of vitamin D), or the extent to which vitamin D alters drug absorption and metabolism, activity or toxicity. Specifically, the review will focus on human studies examining non-calcemic/bone mineralization drug-vitamin D interactions.
Methods
Study selection
A systematic literature search of electronic databases was conducted for articles published through September 1, 2010. Databases that yielded articles meeting the eligibility criteria were: BIOSIS Previews, CAB Abstracts, Cumulative Index of Nursing and Allied Health, Global Health, International Pharmaceutical Abstracts, and Medline.
A search strategy was initially performed using the Medical Subject Headings (MeSH) and keywords “vitamin D”, “cholecalciferol”, “ergocalciferol”, “drug interactions”. Based on articles identified during the initial search, the search terms “colestyramine”, “statin”, “antibiotics”, “cimetidine”, “anticonvulsants”, “glucocorticoids”, “cyclosporins”, “mineral oils”, “hormone replacement therapy”, “weight reduction”, “mineral oils”, “diuretics”, “thiazides”, “hydroxymethylglutaryl-CoA reductase inhibitors”, “histamine H2 antagonists”, “HIV protease inhibitors”, or “immunosuppressive agents” were added in subsequent searches. Additional references within identified primary research or review articles were also examined for eligibility.
Studies were included in the systematic review if they assessed vitamin D intake or concentrations and drug interactions in humans. Reports were excluded if the focus was on vitamin D analogues, osteoporosis or osteopenia treatment, or if vitamin D metabolism was altered as a consequence of the disease process rather than a treatment or an intervention. Animal or cell culture studies were also excluded. Case reports were included for most drug categories, however they were excluded for steroid and antiepileptic drugs because a considerable number of studies with stronger study designs were available for those drug categories.
Data abstraction and quality assessment
Using a standardized data abstraction form, two of the authors (K.R., J.H.R.) abstracted data for each trial. A third author (S.J.O.) reviewed the articles and abstraction forms for accuracy of the classification and quality rating. In cases where the third author disagreed with classification and/or rating assigned by the primary reviewer, the study was discussed among the authors until a consensus was reached. The following information was abstracted from each study: first author, year of publication, location of the study, study design, study population, sample size, duration of participant follow-up, drug dose and formulation, effect on 25(OH)D concentrations or drug level/activity, potential confounders evaluated in the study, and study limitations. If a study reported findings related to both 25(OH)D and 1,25(OH)2D concentrations, only the data related to 25(OH)D were abstracted.
The American Dietetic Association Evidence Analysis classification system and quality criteria checklist 28 was used to assign class and quality ratings. The ADA system was chosen because it is oriented towards medical nutrition interventions and is designed to support translation to clinical practice guidelines. Study classification was based on study design, with randomized controlled trials being assigned a classification of A, cohort studies assigned a B classification, case-control and time series studies assigned a C classification, and cross-sectional, case series, case reports and before-and-after studies assigned a D classification. The quality criteria checklist includes questions in 10 categories relating to the reporting of the research methods and findings: a clear statement of the research question, potential for bias in selection of study participants, comparability of the study groups, methods for handling withdrawals, appropriateness of exposure assessment or the intervention, appropriateness of the outcome assessment, statistical analysis methods, whether the conclusions are supported by the data, and the potential for bias from the study’s funding or sponsorship. Studies that appeared to be free from selection bias, applied appropriate randomization procedures, and had appropriate intervention methods/exposure assessment and outcome measurements received a positive rating. Studies that failed to meet the reporting requirements for six or more of the quality criteria categories received a negative rating. All other studies received a neutral rating.
Results
A total of 1225 reports were identified through the initial search process. Titles were reviewed for eligibility, and 912 manuscripts were excluded at this stage. Abstracts were obtained for the remaining 313 reports. After reviewing the abstracts, 109 unique reports met the full inclusion criteria (Figure 3).
Figure 3.
Flow chart of manuscript identification and inclusion
Included studies are summarized in Supplementary Table 1 (available online). The majority of the studies were classified as Class C (non-randomized trials, case-control studies, or time series; n=30, 28%) or D (cross-sectional, trend, case reports/series, or before-and-after studies; n=69, 63%). Ten of the included studies were randomized controlled trials (RCT, Class A), of which eight were of neutral quality and two were rated as negative quality. None of the included studies were cohort studies (Class B). Only two of the Class C and three of the Class D studies were found to be of positive quality. All positive quality studies were published after 1996, likely reflecting increasing reporting standards for publication.
Drugs that interfere with vitamin D absorption
Bile Acid Sequestrants
The bile acid sequestrants, colestipol and cholestyramine, reduce cholesterol by binding bile acids in the gastrointestinal tract and preventing reabsorption of the bile acids. Bile acid sequestrants may also bind fat-soluble vitamins including vitamin D. As vitamin D metabolites are also present in the bile, increased bile acid excretion could reduce body stores of vitamin D.
Three RCTs (Class A; two neutral quality 29, 30, one negative quality 31), one time series (Class C, negative quality 32), and one before-and-after study (Class D, negative quality33) evaluated the effect of bile acid sequestrants on vitamin D status. One of the RCTs reported a statistically significant decrease in serum 25(OH)D concentrations among children with familial hypercholesterolemia taking 8 g cholestyramine/day for one year compared to controls 30. In contrast, the time series and before-and-after studies reported no significant change from baseline circulating 25(OH)D levels among children taking colestipol for 2–24 months 32, 33. Similarly, two of the RCTs both reported no significant differences in circulating 25(OH)D concentrations between adults taking 24 g cholestyramine/day and a control group after 24 weeks 31 or 7–10 years 29. Overall, these studies suggest that bile acid sequestrants do not alter vitamin D status.
Lipase inhibitors
Orlistat is used as a weight loss aid, and acts by binding the active sites of gastric and pancreatic lipases within the gastrointestinal tract to block absorption of dietary fats, and thus calories 34. As vitamin D is fat soluble, orlistat may also inhibit dietary and supplemental vitamin D absorption 35.
Two RCTs (Class A, both neutral quality 36, 37) and one before-and-after study (Class D, negative quality 38) met the inclusion criteria for this drug category. All three studies reported decreases in 25(OH)D concentrations among participants receiving orlistat. However, in the RCTs, the control groups also experienced a decrease in 25(OH)D concentrations suggesting that the decrease in dietary fat intake may be the reason for the decrease in 25(OH)D concentrations rather than the orlistat itself.
Vitamin D status should be monitored for individuals taking orlistat. If deficient, it would be prudent to recommend that these individuals take vitamin D supplements several hours prior to their orlistat dose to maximize vitamin D absorption.
Drugs that interfere with vitamin D metabolism
Statins
Statins lower serum cholesterol concentrations by inhibiting the rate-limiting enzyme in cholesterol synthesis, HMG Co-A reductase 39. Vitamin D is derived from cholesterol, so by decreasing cholesterol synthesis, statins could also reduce vitamin D synthesis 40, 41. Another potential mechanism for vitamin D-statin interactions is competition for CYP3A4 activity. Atorvastatin, lovastatin and simvastatin are primarily metabolized by CYP3A4 42, 43. Rosuvastatin and fluvastatin are primarily metabolized by CYP2C9 43, 44. Pitavastatin and pravastatin interact minimally with metabolizing enzymes, degrading in the stomach and excreted as parent compound 43, 44.
A total of five studies on statins and vitamin D status, including one RCT (Class A, negative quality 31), one nonrandomized trial (Class C, neutral quality 45) and two before-after studies (Class D, both negative quality 41, with data from one study published in two separate publications 46, 47), and one cross-sectional study (Class D, neutral quality 48) met the inclusion criteria. Three studies reported that atorvastatin therapy increased circulating 25(OH)D 45, 46, 48. One study reported statistically significantly lower concentrations of atorvastatin and its metabolites among participants taking 800 IU/d supplemental vitamin D for 6 weeks compared to those who did not receive supplements (p<0.05) 45. However, cholesterol levels were also lower during vitamin D supplementation despite lower atorvastatin concentrations. The two studies evaluating the effect of pravastatin therapy on 25(OH)D concentrations 31, 41 both reported no significant differences in 25(OH)D concentrations before and after treatment.
Although further study is needed, it appears that only the statins metabolized by CYP3A4 have the potential to interact with vitamin D supplementation. Clinicians should consider whether it is appropriate to ask patients to discontinue vitamin D supplementation while taking atorvastatin, lovastatin or simvastatin, or whether patients should be switched to a different statin in order to continue vitamin D supplementation.
Antimicrobials
Rifampin and isoniazid
Rifampin and isoniazid are used in treating tuberculosis (TB). The complex relationship between vitamin D and TB has long been recognized. Prior to the advent of antibiotics, sun exposure and vitamin D supplements formed the primary treatment for the disease 49. Vitamin D is a modulator of macrophage activity and enhances the production of the antimicrobial protein cathelicidin 50. Vitamin D deficiency has been associated with increased susceptibility to TB infection or reactivation of latent TB infections 51. Treatment with rifampin and isoniazid may also alter vitamin D status, as CYP3A4 is induced by rifampin and inhibited by isoniazid 52.
Six small time series studies (Class C, all negative quality 53–58), each with between 8 and 27 participants, have evaluated the association between rifampin, isoniazid and vitamin D status. Four studies reported that 25(OH)D decreased 53–56, one reported no change 57, and one reported increased 25(OH)D 58 after rifampin and/or isoniazid treatment. Several of the studies noted that the individuals with TB had below normal 25(OH)D concentrations pre-treatment 56, 57. While some of the studies considered the season in which vitamin D status was assessed, few considered dietary or supplemental vitamin D intake, and none assessed UV exposure or stratified by race/skin tone. Thus, it is prudent to monitor 25(OH)D concentrations during rifampin and isoniazid treatment, however if vitamin D deficiency is noted, it may be due to decreased vitamin D exposure rather than a true drug-nutrient interaction.
Hydroxychloroquine
Hydroxychloroquine is used in the treatment of malaria, as well as autoimmune disorders such as systemic lupus erythematosus (SLE). Because individuals with autoimmune diseases often also have photosensitivity and avoid sun exposure, there has been concern that vitamin D deficiency might be common in this population.
One cross-sectional study with comparison group (Class D, neutral quality 59) evaluated the prevalence and predictors of vitamin D deficiency (defined as serum 25(OH)D <10 ng/mL) among 92 adults with SLE. The researchers found that vitamin D deficiency is common among individuals with SLE (n=69, 75%), and individuals taking hydroxychloroquine had higher 25(OH)D concentrations compared to those who were not taking hydroxychloroquine, which the authors hypothesized may be due to a decreased rate of conversion of 25(OH)D to 1,25(OH)2D 59.
Antiepileptic drugs
Physicians have long noted a higher incidence of osteopenia and osteporosis among patients on antiepileptic drugs (AEDs), however the mechanism by which this occurs is not entirely clear. Cell culture studies have shown that phenobarbital (PB), phenytoin (PHT), primidone (PRM), carbamazepine (CBZ), oxcarbazepine and felbamate induce CYP3A4 expression, whereas ethosuximide (ETHS), valproic acid (VPA), and lamotrigine (LTG) have no effect on CYP3A4 activity 60, 61. PB and PHT have also been found to increase CYP24A expression 61, 62, which could result in decreased clearance of vitamin D metabolites and lower serum 25(OH)D levels.
In total, 46 studies have evaluated the effect of AEDs on vitamin D status, however most were small single-institution reports, and only 4 of these studies included more than 100 participants 63–66. The majority of included studies were cross-sectional with (Class D; one positive quality 67, 17 neutral quality 63, 68–83, and 13 negative quality 64–66, 84–93) or without a comparison group (Class D; three neutral quality 94–96, and two negative quality 97, 98). The ten remaining studies were seven time series studies with (Class C; three neutral quality 99–101, two negative quality 102, 103) or without comparison groups (Class C; two neutral quality 104, 105), and three before-and-after studies (Class D; one positive quality 106, one neutral quality 107, and one negative quality 108).
Study design limitations likely contributed to variation in the findings across the 46 studies. Most of the studies that compared AED users to non-AED users found AED use to be associated with lower serum 25(OH)D concentrations 63, 64, 66, 68–74, 76–78, 81, 84, 86, 88–90, 93, 99, 100, 102, 109, however two of these studies reported that the difference in 25(OH)D concentrations between AED users and controls occurred only in the winter months 76, 102. Seven studies reported no significant differences in 25(OH)D concentrations between AED users and non-users 64, 65, 79, 80, 82, 85, 91, 101. Most of the participants in these studies were ambulatory rather than institutionalized AED users, and two of the studies were conducted in lower latitude countries 82, 85. Unexpectedly, one study reported that the individuals on AEDs had higher 25(OH)D concentrations compared to the controls 92, which the authors attributed to adequate sun exposure given that the study participants lived in Florida.
Many studies combined the data for individuals who were on a variety of different single or multidrug AED regimens, and did not adjust for dose or duration of AED use. Of the few studies that reported the effects of specific AEDs on 25(OH)D concentrations, no statistically significant differences in 25(OH)D concentrations were observed between those on the CYP3A4 inducing AEDs compared to normal controls 66, or within individuals over time 107. One study reported no statistically significant difference in 25(OH)D concentrations among individuals on CYP3A4-inducing AEDs compared to those on other AEDs 96. Overall, the literature suggests that the effect of AEDs on vitamin D status may only be evident among individuals with insufficient exposure to exogenous sources of vitamin D (diet, supplements or UV exposure).
Corticosteroids
Glucocorticoids, such as prednisone, hydrocortisone and dexamethasone, are used pharmacologically for a variety of clinical applications including adrenal replacement, immune suppression, and chemotherapy. However, osteoporosis is a well-known complication of corticosteroid therapy. Alterations in vitamin D metabolism have been investigated as a possible mechanism.
Two RCTs (Class A, both neutral quality 110, 111), four time-series (Class C; three neutral quality 112–114, one positive quality 115) and five cross-sectional studies (Class D; one negative 116, three neutral 117–119 and one positive quality 120) have evaluated the effect of prednisone therapy on 25(OH)D concentrations. The majority found no difference in 25(OH)D concentrations in comparison to either pre-treatment concentrations or to a control group 111–113, 116, 117, 119. Lems et al 114 reported that 25(OH)D concentrations decreased after low dose prednisone treatment among healthy controls, which they attributed to seasonal effects given that the study concluded in the fall. In a study of 50 adult rheumatoid arthritits (RA) patients on low dose prednisone, Lund et al 118 also found that 25(OH)D concentrations were significantly lower than the laboratory’s normal values, although none of the study participants were considered deficient. The decreased concentrations may be explained by the fact that the study participants were likely older than the subjects used to establish the laboratory norms. The authors also appropriately note that photosensitivity is a common complication of glucocorticoids and other RA treatments, and the study participants may have been more likely than the general population to avoid sun exposure.
Two studies of prednisolone, one RCT (Class A, neutral quality 121) and one time-series (Class C, neutral quality 122) both found no statistically significant differences in 25(OH)D concentrations pre- vs. post-treatment Six studies, all cross-sectional (Class D; one negative 123, five neutral quality 124–128) did not specify the type of glucocorticoid that the participant received. One study comparing 31 adult RA patients on corticosteroids for at least six months (2.5–10 mg prednisone equivalents/day) to 38 healthy controls found that the corticosteroid users had significantly lower 25(OH)D concentrations compared to healthy controls 124. Similarly, two studies of children and young adults found that individuals with low 25(OH)D concentrations had significantly higher lifetime cumulative glucocorticoid exposure compared to those with higher 25(OH)D concentrations 125, 126. However, the remaining three studies, one in adults 123 and two in children 127, 128, found no significant differences in 25(OH)D concentrations between individuals receiving glucocorticoids and controls or laboratory normal values.
Overall, the studies evaluating the effect of glucocorticoids on vitamin D status suggest that 25(OH)D concentrations are not significantly affected by glucocorticoids, and that the observed association with osteoporosis/osteopenia may be related to drug effects on other parameters of bone metabolism 113. Few of these studies considered potential differences in the glucocorticoid-vitamin D association by body composition, dietary or supplemental vitamin D intake, or UV exposure.
Immunosuppressive agents
Immunosuppressive agents, such as cyclosporine and tacrolimus inhibit T-cell activation, and are used to decrease the risk of rejection of the transplanted tissue following solid organ and hematopoietic cell transplantation. Lower doses of these drugs are also used to treat autoimmune disorders. Osteoporosis is a common long-term side effect, especially among transplant patients who often receive both immunosuppressive agents and steroids.
Cyclosporine
Data from cell culture and animal models indicate that cyclosporine inhibits CYP27A1 129–133 and decreases expression of the vitamin D receptor (VDR) and CYP24 133, which would suggest that cyclosporine could alter circulating 25(OH)D concentrations. One RCT (Class A, neutral quality 134) and five time series studies (Class C; one positive quality 135, one neutral quality 136, and three negative quality 137–139) evaluated the effect of cyclosporine on vitamin D status. None of the studies reported significant differences in 25(OH)D concentrations when comparing the effect of cyclosporine alone or in combination with prednisone.
Tacrolimus
Tacrolimus is metabolized by CYP3A4 and CYP3A5 140, and thus may also be associated with altered 25(OH)D concentrations. One time series study (Class C, negative quality) evaluated the effect of tacrolimus on vitamin D status in individuals who had undergone renal transplantation 139. Again, 25(OH)D concentrations were not significantly different than those of the healthy control group at any of the study time points.
While it does not appear that cyclosporine or tacrolimus alter vitamin D status, osteopenia and osteoporosis are common among this patient population. Thus, it is prudent to monitor vitamin D concentrations in individuals receiving these drugs, and provide supplements as needed to maintain adequate 25(OH)D concentrations. It is likely that the underlying disease state or factors associated with treatment may keep individuals from obtaining adequate vitamin D exposure from sunlight, diet or supplements, rather than a true effect of the immunosuppressant itself on vitamin D status.
Chemotherapeutic agents
A number of chemotherapeutic agents are metabolized by CYP3A4, including etoposide, epipodophyllotoxin, cyclosphosphamide, ifosfamide, vincristine, vinblastine, paclitasel, docetaxel, irinotecan, tamoxifen and imatinib 141, and thus may interact with vitamin D. However, few have been extensively studied with respect to their effect on vitamin D status to date.
Two time series studies (Class C; one neutral quality 142 and one negative quality 143) and one cross-sectional study (Class D, neutral quality 144) evaluated vitamin D status during chemotherapy. All three studies reported no significant changes in 25(OH)D concentrations during treatment of breast, ovarian, uterine, or colorectal cancers with a number of different chemotherapeutic agents (cisplatin, 5-fluorouracil, epirubicin, irinotecan, oxaliplatin, capecitabine, and several monoclonal antibodies). Given the small number of study participants in the studies to date, and the large number of different (often multi-agent) regimens used for cancer treatment, further research is needed. However, because of the high likelihood of vitamin D deficiency due to suboptimal dietary/supplemental intake and decreased UV exposure, vitamin D status should be monitored regularly for patients undergoing cancer treatment.
Highly active antiretroviral agents (HAART)
Highly active antiretroviral therapy (HAART) are a broad category of antiretroviral drugs that inhibit various stages of the human immunodeficiency virus (HIV) life-cycle, and include nucleoside reverse transcriptase inhibitors (NRTI), nucleotide reverse transcriptase inhibitors (NtRTI), non-nucleoside reverse transcriptase inhibitors (NNRTI), protease inhibitors (PI), and entry inhibitors 145. In vitro studies have indicated that HAARTs are metabolized by CYP3A4, and can either induce or inhibit CYP3A4 activity 146, and thus drug-induced induction or inhibition of CYP3A4 could alter rates of 25(OH)D synthesis and degradation. Cozzolino et al 147 reported that conversion of vitamin D3 to 25(OH)D and 1,25(OH)2D, and degradation of the 1,25(OH)2D metabolite was inhibited in human hepatocyte cell cultures exposed to PIs. However, evidence of HAART inhibiting vitamin D bioactivation in humans is currently limited, and inconclusive.
Three cross-sectional studies (Class D; two neutral quality 148, 149, one negative quality 150) met the inclusion criteria for this drug category. The two Spanish studies reported lower serum 25(OH)D concentrations among individuals on HAART compared to those who were not on HAART 148, 150, but the difference was only statistically significant in one study 148. The other study 149 reported that half of the 44 study participants on HAART had deficient 25(OH)D levels (< 34 ng/dL), but this study did not include a non-HAART comparison group.
Given the in vitro data suggesting that vitamin D status might be effected by HAART medications, vitamin D status should be monitored in individuals receiving HAART. Future research in this area should consider body composition changes as a potential covariate effecting vitamin D status. Lipodystrophy, a well described side effect of HAART characterized by alterations in adipose tissue deposition, may also contribute to alterations in circulating 25(OH)D concentrations.
Histamine H2-receptor antagonists
The histamine H2-receptor antagonist, cimetidine, inhibits gastric acid secretion by inhibiting histamine stimulation of the gastric parietal cells. However, animal data shows that cimetidine also inhibits CYP enzymes, including the 25-hydroxylases 151, 152. One time series study (Class C, neutral quality) of nine adults with gastric ulcers found no significant change from baseline serum 25(OH)D concentrations while participants were taking cimetidine, yet serum 25(OH)D concentrations rose significantly once cimetidine was discontinued 153. Without a placebo control or other similar studies published, this finding must be interpreted with caution. Ranitidine, another histamine H2-receptor antagonist, has not been shown to interact with the CYP enzymes in animal models 154.
Drug-vitamin D interactions that induce side effects
Thiazides
Thiazide diuretics are prescribed to reduce blood pressure, treat edema or fluid retention, treat diabetes insipidus, or prevent kidney stones in patients with hypercalciuria. Thiazides reduce the reabsorption of electrolytes from the renal tubules, increase the excretion of electrolytes and fluid, and reduce the excretion of calcium. The combination of thiazide diuretics (decreases urinary calcium excretion) and vitamin D supplementation (enhances intestinal calcium absorption) may theoretically cause or exacerbate hypercalcemia 155.
Excluding reports of patients with altered calcium metabolism due to idiopathic osteoporosis or hyperparathyroidism156–159, three cases of hypercalcemia while on thiazides have been reported in two published manuscripts (Class D; one positive quality160, one negative quality161 ), including: a 78 year old woman taking vitamin D2 (50,000 IU/day), calcium carbonate (1.5 g elemental calcium/day) and hydrochlorothiazide (25 mg/day) 160; an 87 year old woman taking vitamin D (dose not specified), calcium carbonate antacids (1.9 g elemental calcium/day) along with hydrochlorothiazide (50 mg/day) 161; and an 88 year old woman taking vitamin D (1000 IU/day) and calcium carbonate antacids (3.8 g elemental calcium/day) along with hydrochlorothiazide (50 mg/day) 161. These cases were reversible after rehydration and withdrawing the calcium and vitamin D supplementation and the thiazide diuretic. Clinicians should be aware that the combination of thiazide diuretics and vitamin D supplementation may cause hypercalcemia, especially in at-risk individuals, such as the elderly, and individuals with compromised renal function or hyperparathyroidism.
Four additional reports evaluated the effect of thiazide diuretics on serum 25(OH)D concentrations, including one RCT (Class A, negative quality 162), one non-randomized crossover trial (Class C, negative quality 163), and one before-after study 164 and one cross-sectional study 165 (both Class D, negative quality). None of the studies reported significant alterations in 25(OH)D concentrations as a result of thiazide treatment.
Discussion
This systematic review found insufficient evidence to determine whether lipase inhibitors, antimicrobial agents, antiepileptic drugs, HAART or H2 receptor antagonists alter serum 25(OH)D concentrations. Atorvastatin appears to increase 25(OH)D concentrations, while concurrent vitamin D supplementation decreases concentrations of atorvastatin. Use of thiazide diruetics in combination with calcium and vitamin D supplements may induce hypercalcemia in the elderly, or those with compromised renal function or hyperparathyroidism.
The area of drug-vitamin D interactions is a clear example of a situation where lack of evidence does not equate to “no harm”. The available research to date has primarily focused on drugs that are commonly associated with osteoporosis (suggesting a potential effect on vitamin D metabolism), or where case reports of adverse outcomes have been reported in the medical literature. Recent advances in understanding the mechanistic details of CYP3A4 mediated drug metabolism, and a growing appreciation of the role of vitamin D in CYP3A4 expression will likely lead to a systematic evaluation of potential interactions among drugs that are metabolized by CYP3A4, as well as those metabolized by CYP2R1, CYP27A, CYP27B and CYP24.
There is also a need for further research to understand the impact of drugs that inhibit CYP enzyme activity related to vitamin D status. For example, synthetic azole drugs, such as the antimicrobial agent ketoconazole and proton pump inhibitor omeprazole, have been shown to inhibit both CYP3A4 166, 167 and CYP24 168 in vitro, yet no studies to date have evaluated the effect of these drugs on human vitamin D status.
The currently available literature on drug-vitamin D interactions has a number of limitations, as reflected in the number of neutral and negative quality ratings assigned in this review. Much of the literature to date is based on small case-control studies, case studies, or secondary analyses of clinical data collected for other reasons. Many of the studies were hospital-based and lacked relevant comparison groups. Most studies failed to evaluate dietary or supplemental vitamin D intake and sun exposure, as potential effect modifiers. And very few studies considered body weight or composition as a potential confounder effecting both vitamin D concentrations and drug response. The majority of studies also lacked statistical power to adjust for appropriate covariates or rule out false negative findings. For many of the studies where individuals taking a drug were found to have lower 25(OH)D levels than a non-drug comparison group, the lack of data collection on vitamin D intake and UV exposure makes it difficult to determine whether the observed vitamin D deficiency is due to insufficient intake or due to the drug itself.
Because vitamin D is highly hydrophobic and has several metabolites, serum vitamin D determinations are technically challenging. Methodology for assessing vitamin D status has improved significantly in recent years, and the older data reported in many of the studies included in this systematic review may not be accurate or comparable to more current data. Currently, high performance liquid chromatography (HPLC) or liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) is considered the gold standard technique, although when performed by experienced users, radioimmunoassay (RIA) techniques correlate very closely with LC-MS/MS 169. Commercially available testing kits have been found to produce highly variable results when performed by inexperienced users 170. As a result of regional surveys revealing significant variation between laboratories, an international standardization group, the vitamin D External Quality Assessment Scheme (DEQAS), was started in 1989 171. In 2009, the US National Institute of Standards and Technology (NIST) developed a vitamin D standard (standard reference material 972, Vitamin D in Human Serum) with certified and reference values for 25(OH)D2, 25(OH)D3, and 3-epi-25(OH)D3 172. Supplies of this standard quickly sold out, and NIST does not plan to continue producing this standard due to difficulties in formulating the product. A companion NIST product, SRM 2972, is a set of ethanol-based calibration solutions and has certified values for 25(OH)D2 and 25(OH)D3 which is currently available. NIST has also established a Vitamin D Metabolites Quality Assurance Program (VitDQAP, http://www.nist.gov/mml/analytical/vitdqap.cfm) in collaboration with the National Institutes of Health (NIH) Office of Dietary Supplements (ODS).
Given the increasing prevalence of vitamin D supplementation in the general population, continued evaluation of potential drug-vitamin D interactions is warranted. Larger studies with stronger study designs are needed to clarify potential drug-vitamin D interactions. Future research in this area should address the limitations identified in this review, specifically with prospective data collection including assessment of vitamin D exposure and potential confounding factors such as body weight/composition and seasonality/UV exposure. Future studies should also use standardized vitamin D assay methodologies in a laboratory that participates in external quality assessment protocols specific to vitamin D. Until further research is available, health care professionals should be aware of the potential for drug-vitamin D interactions, assess their clients’ use of dietary supplements, and monitor serum 25(OH)D concentrations where indicated with the ultimate goal of achieving adequate serum 25(OH)D concentrations while optimizing drug efficacy and minimizing drug toxicity.
Supplementary Material
Acknowledgments
Funding: Support for S.J.O. was provided by training grant T32 CA132670 from the National Cancer Institute.
Contributor Information
Kim Robien, Associate Professor, Division of Epidemiology and Community Health; and Member, Cancer Outcomes and Survivorship Program, Masonic Cancer Center, University of Minnesota 1300 S. Second St., Suite 300, Minneapolis, MN 55454, Phone: 612-625-8279, Fax: 612-624-0315.
Sarah J. Oppeneer, Doctoral Student, Division of Epidemiology and Community Health, University of Minnesota, 1300 S. Second St., Suite 300, Minneapolis MN 55454, Phone: 612-625-4542, Fax: 612-624-0315.
Julia A. Kelly, Librarian, University Libraries, University of Minnesota, 1984 Buford Ave, St. Paul, MN 55108, Phone: 612-624-4781.
Jill M. Hamilton-Reeves, Assistant Professor, University of Kansas Medical Center, School of Health Professions, Department of Dietetics & Nutrition, 3901 Rainbow Boulevard, Mail Stop 4019, 4093 DELP, Kansas City, KS 66160, Phone: 913-588-7650, Fax: 913-588-8946.
References
- 1.Cohen-Lahav M, Douvdevani A, Chaimovitz C, Shany S. The anti-inflammatory activity of 1,25-dihydroxyvitamin D3 in macrophages. J Steroid Biochem Mol Biol. 2007 Mar;103(3–5):558–562. doi: 10.1016/j.jsbmb.2006.12.093. [DOI] [PubMed] [Google Scholar]
- 2.Boucher BJ. Vitamin D insufficiency and diabetes risks. Curr Drug Targets. 2011 Jan;12(1):61–87. doi: 10.2174/138945011793591653. [DOI] [PubMed] [Google Scholar]
- 3.World Health Organization. Working Group Report 5. Vol. 5. Lyon, France: 2008. International Agency for Research on Cancer (IARC). Vitamin D and Cancer. [Google Scholar]
- 4.Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C. Vitamin D: modulator of the immune system. Curr Opin Pharmacol. 2010 Aug;10(4):482–496. doi: 10.1016/j.coph.2010.04.001. [DOI] [PubMed] [Google Scholar]
- 5.Reddy Vanga S, Good M, Howard PA, Vacek JL. Role of vitamin D in cardiovascular health. Am J Cardiol. 2010 Sep 15;106(6):798–805. doi: 10.1016/j.amjcard.2010.04.042. [DOI] [PubMed] [Google Scholar]
- 6.Vitamin D sales strong in 2010, supply costs rising. Nutrition Business Journal. 2011;16 [Google Scholar]
- 7.Moshfegh A, Goldman J, Ahuja J, Rhodes D, LaComb R. What We Eat In America, NHANES 2005–2006: Usual nutrient intake from food and water compared to 1997 Dietary Reference Intakes for vitamin D, calcium, phosphorus and magnesium. Beltsville, MD: US Department of Agriculture, Agricultural Research Service; 2008. [Google Scholar]
- 8.Office of Dietary Supplements, National Institutes of Health. [Accessed November 17, 2011];Dietary Supplement Fact Sheet: Vitamin D. http://ods.od.nih.gov/factsheets/VitaminD-HealthProfessional#h9.
- 9.Henry HL. The 25-Hydroxyvitamin D 1alpha-hydroxylase. In: Feldman D, Pike JW, Gloriueux FH, editors. Vitamin D. 2. Vol. 1. Boston: Academic Press; 2005. pp. 69–83. [Google Scholar]
- 10.Norman AW, Mizwicki MT, Norman DP. Steroid-hormone rapid actions, membrane receptors and a conformational ensemble model. Nat Rev Drug Discov. 2004 Jan;3(1):27–41. doi: 10.1038/nrd1283. [DOI] [PubMed] [Google Scholar]
- 11.Pascussi JM, Gerbal-Chaloin S, Drocourt L, Maurel P, Vilarem MJ. The expression of CYP2B6, CYP2C9 and CYP3A4 genes: a tangle of networks of nuclear and steroid receptors. Biochim Biophys Acta. 2003 Feb 17;1619(3):243–253. doi: 10.1016/s0304-4165(02)00483-x. [DOI] [PubMed] [Google Scholar]
- 12.Chatterjee B, Echchgadda I, Song CS. Vitamin D receptor regulation of the steroid/bile acid sulfotransferase SULT2A1. Methods in enzymology. 2005;400:165–191. doi: 10.1016/S0076-6879(05)00010-8. [DOI] [PubMed] [Google Scholar]
- 13.Fan J, Liu S, Du Y, Morrison J, Shipman R, Pang KS. Up-regulation of transporters and enzymes by the vitamin D receptor ligands, 1alpha,25-dihydroxyvitamin D3 and vitamin D analogs, in the Caco-2 cell monolayer. J Pharmacol Exp Ther. 2009 Aug;330(2):389–402. doi: 10.1124/jpet.108.149815. [DOI] [PubMed] [Google Scholar]
- 14.Institute of Medicine. Dietary Reference Intakes for Vitamin D and Calcium. Vol. 2010. Washington, D.C: Institute of Medicine; 2011. [Google Scholar]
- 15.Bischoff-Ferrari HA, Giovannucci E, Willett WC, Dietrich T, Dawson-Hughes B. Estimation of optimal serum concentrations of 25-hydroxyvitamin D for multiple health outcomes. Am J Clin Nutr. 2006 Jul;84(1):18–28. doi: 10.1093/ajcn/84.1.18. [DOI] [PubMed] [Google Scholar]
- 16.Vieth R, Bischoff-Ferrari H, Boucher BJ, et al. The urgent need to recommend an intake of vitamin D that is effective. Am J Clin Nutr. 2007 Mar;85(3):649–650. doi: 10.1093/ajcn/85.3.649. [DOI] [PubMed] [Google Scholar]
- 17.Hollis BW. Circulating 25-hydroxyvitamin D levels indicative of vitamin D sufficiency: implications for establishing a new effective dietary intake recommendation for vitamin D. J Nutr. 2005 Feb;135(2):317–322. doi: 10.1093/jn/135.2.317. [DOI] [PubMed] [Google Scholar]
- 18.Wortsman J, Matsuoka LY, Chen TC, Lu Z, Holick MF. Decreased bioavailability of vitamin D in obesity. Am J Clin Nutr. 2000 Sep;72(3):690–693. doi: 10.1093/ajcn/72.3.690. [DOI] [PubMed] [Google Scholar]
- 19.Webb AR, Kline L, Holick MF. Influence of season and latitude on the cutaneous synthesis of vitamin D3: exposure to winter sunlight in Boston and Edmonton will not promote vitamin D3 synthesis in human skin. J Clin Endocrinol Metab. 1988 Aug;67(2):373–378. doi: 10.1210/jcem-67-2-373. [DOI] [PubMed] [Google Scholar]
- 20.Dawson-Hughes B, Harris SS, Dallal GE. Plasma calcidiol, season, and serum parathyroid hormone concentrations in healthy elderly men and women. Am J Clin Nutr. 1997 Jan;65(1):67–71. doi: 10.1093/ajcn/65.1.67. [DOI] [PubMed] [Google Scholar]
- 21.Dawson-Hughes B, Heaney RP, Holick MF, Lips P, Meunier PJ, Vieth R. Estimates of optimal vitamin D status. Osteoporos Int. 2005 Jul;16(7):713–716. doi: 10.1007/s00198-005-1867-7. [DOI] [PubMed] [Google Scholar]
- 22.Luo G, Guenthner T, Gan LS, Humphreys WG. CYP3A4 induction by xenobiotics: biochemistry, experimental methods and impact on drug discovery and development. Curr Drug Metab. 2004 Dec;5(6):483–505. doi: 10.2174/1389200043335397. [DOI] [PubMed] [Google Scholar]
- 23.Zhou SF. Drugs behave as substrates, inhibitors and inducers of human cytochrome P450 3A4. Curr Drug Metab. 2008 May;9(4):310–322. doi: 10.2174/138920008784220664. [DOI] [PubMed] [Google Scholar]
- 24.Watkins PB, Wrighton SA, Schuetz EG, Molowa DT, Guzelian PS. Identification of glucocorticoid-inducible cytochromes P-450 in the intestinal mucosa of rats and man. J Clin Invest. 1987 Oct;80(4):1029–1036. doi: 10.1172/JCI113156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kolars JC, Schmiedlin-Ren P, Schuetz JD, Fang C, Watkins PB. Identification of rifampin-inducible P450IIIA4 (CYP3A4) in human small bowel enterocytes. J Clin Invest. 1992 Nov;90(5):1871–1878. doi: 10.1172/JCI116064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Thummel KE, Brimer C, Yasuda K, et al. Transcriptional control of intestinal cytochrome P-4503A by 1alpha,25-dihydroxy vitamin D3. Mol Pharmacol. 2001 Dec;60(6):1399–1406. doi: 10.1124/mol.60.6.1399. [DOI] [PubMed] [Google Scholar]
- 27.Lindh JD, Andersson ML, Eliasson E, Bjorkhem-Bergman L. Seasonal variation in blood drug concentrations and a potential relationship to vitamin D. Drug Metab Dispos. 2011 May;39(5):933–937. doi: 10.1124/dmd.111.038125. [DOI] [PubMed] [Google Scholar]
- 28.American Dietetic Association. Evidence Analysis Manual: Steps in the ADA Evidence Analysis Process. Chicago, IL: American Dietetic Association; 2008. [Google Scholar]
- 29.Hoogwerf BJ, Hibbard DM, Hunninghake DB. Effects of long-term cholestyramine administration on vitamin D and parathormone levels in middle-aged men with hypercholesterolemia. J Lab Clin Med. 1992 Apr;119(4):407–411. [PubMed] [Google Scholar]
- 30.Tonstad S, Knudtzon J, Sivertsen M, Refsum H, Ose L. Efficacy and safety of cholestyramine therapy in peripubertal and prepubertal children with familial hypercholesterolemia. J Pediatr. 1996 Jul;129(1):42–49. doi: 10.1016/s0022-3476(96)70188-9. [DOI] [PubMed] [Google Scholar]
- 31.Ismail F, Corder CN, Epstein S, Barbi G, Thomas S. Effects of pravastatin and cholestyramine on circulating levels of parathyroid hormone and vitamin D metabolites. Clin Ther. 1990 Sep-Oct;12(5):427–430. [PubMed] [Google Scholar]
- 32.Tsang RC, Roginsky MS, Mellies MJ, Glueck CJ. Plasma 25-hydroxy-vitamin D in familial hypercholesterolemic children receiving colestipol resin. Pediatr Res. 1978;12(10):980–982. doi: 10.1203/00006450-197810000-00006. [DOI] [PubMed] [Google Scholar]
- 33.Schwarz KB, Goldstein PD, Witztum JL, Schonfeld G. Fat-soluble vitamin concentrations in hypercholesterolemic children treated with colestipol. Pediatrics. 1980 Feb;65(2):243–250. [PubMed] [Google Scholar]
- 34.Reitsma JB, Castro Cabezas M, de Bruin TW, Erkelens DW. Relationship between improved postprandial lipemia and low-density lipoprotein metabolism during treatment with tetrahydrolipstatin, a pancreatic lipase inhibitor. Metab Clin Exp. 1994 Mar;43(3):293–298. doi: 10.1016/0026-0495(94)90095-7. [DOI] [PubMed] [Google Scholar]
- 35.Guerciolini R. Mode of action of orlistat. Int J Obes Relat Metab Disord. 1997 Jun;21(Suppl 3):S12. [PubMed] [Google Scholar]
- 36.Gotfredsen A, Westergren Hendel H, Andersen T. Influence of orlistat on bone turnover and body composition. Int J Obes Relat Metab Disord. 2001 Aug;25(8):1154–1160. doi: 10.1038/sj.ijo.0801639. [DOI] [PubMed] [Google Scholar]
- 37.James WP, Avenell A, Broom J, Whitehead J. A one-year trial to assess the value of orlistat in the management of obesity. Int J Obes Relat Metab Disord. 1997 Jun;21(Suppl 3):S24–30. [PubMed] [Google Scholar]
- 38.McDuffie JR, Calis KA, Booth SL, Uwaifo GI, Yanovski JA. Effects of orlistat on fat-soluble vitamins in obese adolescents. Pharmacotherapy. 2002;7(1):814–822. doi: 10.1592/phco.22.11.814.33627. [DOI] [PubMed] [Google Scholar]
- 39.Istvan ES, Deisenhofer J. Structural mechanism for statin inhibition of HMG-CoA reductase. Science. 2001 May 11;292(5519):1160–1164. doi: 10.1126/science.1059344. [DOI] [PubMed] [Google Scholar]
- 40.Dobs AS, Levine MA, Margolis S. Effects of pravastatin, a new HMG-CoA reductase inhibitor, on vitamin D synthesis in man. Metabolism. 1991 May;40(5):524–528. doi: 10.1016/0026-0495(91)90235-o. [DOI] [PubMed] [Google Scholar]
- 41.Montagnani M, Lore F, Di Cairano G, et al. Effects of pravastatin treatment on vitamin D metabolites. Clin Ther. 1994 Sep-Oct;16(5):824–829. [PubMed] [Google Scholar]
- 42.Vaughan CJ, Gotto AM., Jr Update on statins: 2003. Circulation. 2004 Aug 17;110(7):886–892. doi: 10.1161/01.CIR.0000139312.10076.BA. [DOI] [PubMed] [Google Scholar]
- 43.Williams D, Feely J. Pharmacokinetic-pharmacodynamic drug interactions with HMG-CoA reductase inhibitors. Clin Pharmacokinet. 2002;41(5):343–370. doi: 10.2165/00003088-200241050-00003. [DOI] [PubMed] [Google Scholar]
- 44.Neuvonen PJ, Niemi M, Backman JT. Drug interactions with lipid-lowering drugs: mechanisms and clinical relevance. Clin Pharmacol Ther. 2006 Dec;80(6):565–581. doi: 10.1016/j.clpt.2006.09.003. [DOI] [PubMed] [Google Scholar]
- 45.Schwartz JB. Effects of vitamin D supplementation in atorvastatin-treated patients: a new drug interaction with an unexpected consequence. Clin Pharmacol Ther. 2009 Feb;85(2):198–203. doi: 10.1038/clpt.2008.165. [DOI] [PubMed] [Google Scholar]
- 46.Perez-Castrillon JL, Vega G, Abad L, et al. Effects of atorvastatin on vitamin D levels in patients with acute ischemic heart disease. Amn J Cardiol. 2007;99(7):903–905. doi: 10.1016/j.amjcard.2006.11.036. [DOI] [PubMed] [Google Scholar]
- 47.Perez-Castrillon JL, Abad L, Vega G, et al. Effect of atorvastatin on bone mineral density in patients with acute coronary syndrome. Eur Rev Med Pharmacol Sci. 2008 Mar-Apr;12(2):83–88. [PubMed] [Google Scholar]
- 48.Aloia JF, Li-Ng M, Pollack S. Statins and vitamin D. Am J Cardiol. 2007 Oct 15;100(8):1329. doi: 10.1016/j.amjcard.2007.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gauvain S. Calciferol in the treatment of tuberculous glands. Tubercle. 1948 Nov;29(11):259–264. doi: 10.1016/s0041-3879(48)80036-x. [DOI] [PubMed] [Google Scholar]
- 50.Chocano Bedoya P, Ronnenberg AG. Vitamin D and tuberculosis. Nutr Rev. 2009;67(5):289–293. doi: 10.1111/j.1753-4887.2009.00195.x. [DOI] [PubMed] [Google Scholar]
- 51.Nnoaham KE, Clarke A. Low serum vitamin D levels and tuberculosis: a systematic review and meta-analysis. Int J Epidemiol. 2008 Feb;37(1):113–119. doi: 10.1093/ije/dym247. [DOI] [PubMed] [Google Scholar]
- 52.Zhou S, Chan E, Lim LY, et al. Therapeutic drugs that behave as mechanism-based inhibitors of cytochrome P450 3A4. Curr Drug Metab. 2004 Oct;5(5):415–442. doi: 10.2174/1389200043335450. [DOI] [PubMed] [Google Scholar]
- 53.Brodie MJ, Boobis AR, Dollery CT, et al. Rifampicin and vitamin D metabolism. Clin Pharmacol Ther. 1980 Jun;27(6):810–814. doi: 10.1038/clpt.1980.115. [DOI] [PubMed] [Google Scholar]
- 54.Brodie MJ, Boobis AR, Hillyard CJ, Abeyasekera G, MacIntyre I, Park BK. Effect of isoniazid on vitamin D metabolism and hepatic monooxygenase activity. Clin Pharmacol Ther. 1981 Sep;30(3):363–367. doi: 10.1038/clpt.1981.173. [DOI] [PubMed] [Google Scholar]
- 55.Brodie MJ, Boobis AR, Hillyard CJ, et al. Effect of rifampicin and isoniazid on vitamin D metabolism. Clin Pharmacol Ther. 1982 Oct;32(4):525–530. doi: 10.1038/clpt.1982.197. [DOI] [PubMed] [Google Scholar]
- 56.Davies PD, Brown RC, Church HA, Woodhead JS. The effect of anti-tuberculosis chemotherapy on vitamin D and calcium metabolism. Tubercle. 1987 Dec;68(4):261–266. doi: 10.1016/0041-3879(87)90066-3. [DOI] [PubMed] [Google Scholar]
- 57.Williams SE, Wardman AG, Taylor GA, Peacock M, Cooke NJ. Long term study of the effect of rifampicin and isoniazid on vitamin D metabolism. Tubercle. 1985 Mar;66(1):49–54. doi: 10.1016/0041-3879(85)90053-4. [DOI] [PubMed] [Google Scholar]
- 58.Martinez ME, Gonzalez J, Sanchez-Cabezudo MJ, Pena JM, Vazquez JJ. Remission of hypercalciuria in patients with tuberculosis after treatment. Calcif Tissue Int. 1996 Jul;59(1):17–20. doi: 10.1007/s002239900078. [DOI] [PubMed] [Google Scholar]
- 59.Ruiz-Irastorza G, Egurbide MV, Olivares N, Martinez-Berriotxoa A, Aguirre C. Vitamin D deficiency in systemic lupus erythematosus: prevalence, predictors and clinical consequences. Rheumatology. 2008;47:920–923. doi: 10.1093/rheumatology/ken121. [DOI] [PubMed] [Google Scholar]
- 60.Tanaka E. Clinically significant pharmacokinetic drug interactions between antiepileptic drugs. J Clin Pharm Ther. 1999 Apr;24(2):87–92. doi: 10.1046/j.1365-2710.1999.00201.x. [DOI] [PubMed] [Google Scholar]
- 61.Pascussi JM, Robert A, Nguyen M, et al. Possible involvement of pregnane X receptor-enhanced CYP24 expression in drug-induced osteomalacia. J Clin Invest. 2005 Jan;115(1):177–186. doi: 10.1172/JCI21867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Xu Y, Hashizume T, Shuhart MC, et al. Intestinal and hepatic CYP3A4 catalyze hydroxylation of 1alpha,25-dihydroxyvitamin D(3): implications for drug-induced osteomalacia. Mol Pharmacol. 2006 Jan;69(1):56–65. doi: 10.1124/mol.105.017392. [DOI] [PubMed] [Google Scholar]
- 63.Morijiri Y, Sato T. Factors causing rickets in institutionalised handicapped children on anticonvulsant therapy. Arch Dis Child. 1981 Jun;56(6):446–449. doi: 10.1136/adc.56.6.446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Krause KH, Berlit P, Bonjour JP, Schmidt-Gayk H, Schellenberg B, Gillen J. Vitamin status in patients on chronic anticonvulsant therapy. Int J Vitam Nutr Res. 1982;52(4):375–385. [PubMed] [Google Scholar]
- 65.Gough H, Bissesar A, Goggin T, et al. Factors associated with the biochemical changes in vitamin D and calcium metabolism in institutionalized patients with epilepsy. Ir J Med Sci. 1986 Jun;155(6):181–189. doi: 10.1007/BF02939839. [DOI] [PubMed] [Google Scholar]
- 66.Gough H, Goggin T, Bissessar A, Baker M, Crowley M, Callaghan N. A comparative study of the relative influence of different anticonvulsant drugs, UV exposure and diet on vitamin D and calcium metabolism in out-patients with epilepsy. Q J Med. 1986 Jun;59(230):569–577. [PubMed] [Google Scholar]
- 67.Nettekoven S, Strohle A, Trunz B, et al. Effects of antiepileptic drug therapy on vitamin D status and biochemical markers of bone turnover in children with epilepsy. Eur J Pediatr. 2008;167(12):1369–1377. doi: 10.1007/s00431-008-0672-7. [DOI] [PubMed] [Google Scholar]
- 68.Hahn TJ, Hendin BA, Scharp CR, Haddad JG., Jr Effect of chronic anticonvulsant therapy on serum 25-hydroxycalciferol levels in adults. N Engl J Med. 1972 Nov 2;287(18):900–904. doi: 10.1056/NEJM197211022871803. [DOI] [PubMed] [Google Scholar]
- 69.Bouillon R, Reynaert J, Claes JH, Lissens W, De Moor P. The effect of anticonvulsant therapy on serum levels of 25-hydroxy-vitamin D, calcium, and parathyroid hormone. J Clin Endocrinol Metab. 1975 Dec;41(06):1130–1135. doi: 10.1210/jcem-41-6-1130. [DOI] [PubMed] [Google Scholar]
- 70.Jubiz W, Haussler MR, McCain TA, Tolman KG. Plasma 1,25-dihydroxyvitamin D levels in patients receiving anticonvulsant drugs. J Clin Endocrinol Metab. 1977 Apr;44(4):617–621. doi: 10.1210/jcem-44-4-617. [DOI] [PubMed] [Google Scholar]
- 71.Pylypchuk G, Oreopoulos DG, Wilson DR, et al. Calcium metabolism in adult outpatients with epilepsy receiving long-term anticonvulsant therapy. Can Med Assoc J. 1978 Mar 18;118(6):635–638. [PMC free article] [PubMed] [Google Scholar]
- 72.Weisman Y, Andriola M, Reiter E, Gruskin A, Root A. Serum concentrations of 25-hydroxyvitamin D in Florida children: effect of anticonvulsant drugs. South Med J. 1979 Apr;72(4):400–401. 408. doi: 10.1097/00007611-197904000-00009. [DOI] [PubMed] [Google Scholar]
- 73.Christensen CK, Lund B, Lund BJ, Sorensen OH, Nielsen HE, Mosekilde L. Reduced 2,25-dihydroxyvitamin D and 24,25-dihydroxyvitamin D in epileptic patients receiving chronic combined anticonvulsant therapy. Metab Bone Dis Relat Res. 1981;3(1):17–22. doi: 10.1016/s0221-8747(81)80018-5. [DOI] [PubMed] [Google Scholar]
- 74.Hoikka V, Savolainen K, Alhava EM, Sivenius J, Karjalainen P, Repo A. Osteomalacia in institutionalized epileptic patients on long-term anticonvulsant therapy. Acta Neurol Scand. 1981 Aug;64(2):122–131. doi: 10.1111/j.1600-0404.1981.tb04394.x. [DOI] [PubMed] [Google Scholar]
- 75.Hoikka V, Savolainen K, Karjalainen P, Alhava EM, Sivenius J. Treatment of osteomalacia in institutionalized epileptic patients on long-term anticonvulsant therapy. Ann Clin Res. 1982 Apr;14(2):72–75. [PubMed] [Google Scholar]
- 76.Keck E, Gollnick B, Reinhardt D, Karch D, Peerenboom H, Kruskemper HL. Calcium metabolism and vitamin D metabolite levels in children receiving anticonvulsant drugs. Eur J Pediatr. 1982 Sep;139(1):52–55. doi: 10.1007/BF00442080. [DOI] [PubMed] [Google Scholar]
- 77.Lamberg-Allardt C, Wilska M, Saraste KL, Gronlund T. Vitamin D status of ambulatory and nonambulatory mentally retarded children with and without carbamazepine treatment. Ann Nutr Metab. 1990;34(4):216–220. doi: 10.1159/000177590. [DOI] [PubMed] [Google Scholar]
- 78.Valimaki MJ, Tiihonen M, Laitinen K, et al. Bone mineral density measured by dual-energy x-ray absorptiometry and novel markers of bone formation and resorption in patients on antiepileptic drugs. J Bone Miner Res. 1994 May;9(5):631–637. doi: 10.1002/jbmr.5650090507. [DOI] [PubMed] [Google Scholar]
- 79.Baer MT, Kozlowski BW, Blyler EM, Trahms CM, Taylor ML, Hogan MP. Vitamin D, calcium, and bone status in children with developmental delay in relation to anticonvulsant use and ambulatory status. Am J Clin Nutr. 1997 Apr;65(4):1042–1051. doi: 10.1093/ajcn/65.4.1042. [DOI] [PubMed] [Google Scholar]
- 80.Henderson RC. Vitamin D levels in noninstitutionalized children with cerebral palsy. J Child Neurol. 1997 Oct;12(7):443–447. doi: 10.1177/088307389701200706. [DOI] [PubMed] [Google Scholar]
- 81.Telci A, Cakatay U, Kurt BB, et al. Changes in bone turnover and deoxypyridinoline levels in epileptic patients. Clin Chem Lab Med. 2000 Jan;38(1):47–50. doi: 10.1515/CCLM.2000.008. [DOI] [PubMed] [Google Scholar]
- 82.Filardi S, Guerreiro CA, Magna LA, Marques Neto JF. Bone mineral density, vitamin D and anticonvulsant therapy. Arq Neuropsiquiatr. 2000 Sep;58(3A):616–620. doi: 10.1590/s0004-282x2000000400003. [DOI] [PubMed] [Google Scholar]
- 83.Mintzer S, Boppana P, Toguri J, DeSantis A. Vitamin D levels and bone turnover in epilepsy patients taking carbamazepine or oxcarbazepine. Epilepsia. 2006 Mar;47(3):510–515. doi: 10.1111/j.1528-1167.2006.00460.x. [DOI] [PubMed] [Google Scholar]
- 84.Stamp TC, Round JM, Rowe DJ, Haddad JG. Plasma levels and therapeutic effect of 25-hydroxycholecalciferol in epileptic patients taking anticonvulsant drugs. Br Med J. 1972 Oct 7;4(5831):9–12. doi: 10.1136/bmj.4.5831.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Weisman Y, Fattal A, Eisenberg Z, Harel S, Spirer Z, Harell A. Decreased serum 24,25-dehydroxy vitamin D concentrations in children receiving chronic anticonvulsant therapy. Br Med J. 1979;2(6189):521–523. doi: 10.1136/bmj.2.6189.521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tjellesen L, Christiansen C. Serum vitamin D metabolites in epileptic patients treated with 2 different anti-convulsants. Acta Neurol Scand. 1982 Sep;66(3):335–341. doi: 10.1111/j.1600-0404.1982.tb06853.x. [DOI] [PubMed] [Google Scholar]
- 87.Zerwekh JE, Homan R, Tindall R, Pak CY. Decreased serum 24,25-dihydroxyvitamin D concentration during long-term anticonvulsant therapy in adult epileptics. Ann Neurol. 1982 Aug;12(2):184–186. doi: 10.1002/ana.410120209. [DOI] [PubMed] [Google Scholar]
- 88.Davie MW, Emberson CE, Lawson DE, et al. Low plasma 25-hydroxyvitamin D and serum calcium levels in institutionalized epileptic subjects: associated risk factors, consequences and response to treatment with vitamin D. Q J Med. 1983 Winter;52(205):79–91. [PubMed] [Google Scholar]
- 89.Hoikka V, Alhava EM, Karjalainen P, et al. Carbamazepine and bone mineral metabolism. Acta Neurol Scand. 1984 Aug;70(2):77–80. doi: 10.1111/j.1600-0404.1984.tb00806.x. [DOI] [PubMed] [Google Scholar]
- 90.Rajantie J, Lamberg-Allardt C, Wilska M. Does carbamazepine treatment lead to a need of extra vitamin D in some mentally retarded children? Acta Paediatr Scand. 1984 May;73(3):325–328. doi: 10.1111/j.1651-2227.1994.tb17742.x. [DOI] [PubMed] [Google Scholar]
- 91.Weinstein RS, Bryce GF, Sappington LJ, King DW, Gallagher BB. Decreased serum ionized calcium and normal vitamin D metabolite levels with anticonvulsant drug treatment. J Clin Endocrinol Metab. 1984 Jun;58(6):1003–1009. doi: 10.1210/jcem-58-6-1003. [DOI] [PubMed] [Google Scholar]
- 92.Williams C, Netzloff M, Folkerts L, Vargas A, Garnica A, Frias J. Vitamin D metabolism and anticonvulsant therapy: effect of sunshine on incidence of osteomalacia. South Med J. 1984 Jul;77(7):834–836. 842. doi: 10.1097/00007611-198407000-00011. [DOI] [PubMed] [Google Scholar]
- 93.Nishiyama S, Kuwahara T, Matsuda I. Decreased bone density in severely handicapped children and adults, with reference to the influence of limited mobility and anticonvulsant medication. Eur J Pediatr. 1986 Feb;144(5):457–463. doi: 10.1007/BF00441738. [DOI] [PubMed] [Google Scholar]
- 94.Fogelman I, Gray JM, Gardner MD, et al. Do anticonvulsant drugs commonly induce osteomalacia? Scott Med J. 1982 Apr;27(2):136–142. doi: 10.1177/003693308202700205. [DOI] [PubMed] [Google Scholar]
- 95.Guo CY, Ronen GM, Atkinson SA. Long-term valproate and lamotrigine treatment may be a marker for reduced growth and bone mass in children with epilepsy. Epilepsia. 2001 Sep;42(9):1141–1147. doi: 10.1046/j.1528-1157.2001.416800.x. [DOI] [PubMed] [Google Scholar]
- 96.Farhat G, Yamout B, Mikati MA, Demirjian S, Sawaya R, El-Hajj Fuleihan G. Effect of antiepileptic drugs on bone density in ambulatory patients. Neurology. 2002 May 14;58(9):1348–1353. doi: 10.1212/wnl.58.9.1348. [DOI] [PubMed] [Google Scholar]
- 97.Marcus JC, Pettifor JM. Folate and mineral metabolism in poorly nourished epileptic children. Arch Neurol. 1980 Dec;37(12):772–774. doi: 10.1001/archneur.1980.00500610052008. [DOI] [PubMed] [Google Scholar]
- 98.Maeda K, Ikeda H. High 1,25-dihydroxyvitamin D and low 25-hydroxyvitamin D concentrations in plasma in patients receiving antiepileptic drugs. Jpn J Psychiatry Neurol. 1986 Mar;40(1):57–60. doi: 10.1111/j.1440-1819.1986.tb01612.x. [DOI] [PubMed] [Google Scholar]
- 99.Winnacker JL, Yeager H, Saunders JA, Russell B, Anast CS. Rickets in children receiving anticonvulsant drugs. Biochemical and hormonal markers. Am J Dis Child. 1977 Mar;131(3):286–290. doi: 10.1001/archpedi.1977.02120160040005. [DOI] [PubMed] [Google Scholar]
- 100.Markestad T, Ulstein M, Strandjord RE, Aksnes L, Aarskog D. Anticonvulsant drug therapy in human pregnancy: effects on serum concentrations of vitamin D metabolites in maternal and cord blood. Am J Obstet Gynecol. 1984 Oct 1;150(3):254–258. doi: 10.1016/s0002-9378(84)90361-2. [DOI] [PubMed] [Google Scholar]
- 101.Ala-Houhala M, Korpela R, Koivikko M, Koskinen T, Koskinen M, Koivula T. Long-term anticonvulsant therapy and vitamin D metabolism in ambulatory pubertal children. Neuropediatrics. 1986 Nov;17(4):212–216. doi: 10.1055/s-2008-1052532. [DOI] [PubMed] [Google Scholar]
- 102.Riancho JA, del Arco C, Arteaga R, Herranz JL, Albajar M, Macias JG. Influence of solar irradiation on vitamin D levels in children on anticonvulsant drugs. Acta Neurol Scand. 1989 Apr;79(4):296–299. doi: 10.1111/j.1600-0404.1989.tb03788.x. [DOI] [PubMed] [Google Scholar]
- 103.Nicolaidou P, Georgouli H, Kotsalis H, et al. Effects of anticonvulsant therapy on vitamin D status in children: prospective monitoring study. J Child Neurol. 2006 Mar;21(3):205–209. doi: 10.2310/7010.2006.00050. [DOI] [PubMed] [Google Scholar]
- 104.Bramswig S, Zittermann A, Berthold HK. Carbamazepine does not alter biochemical parameters of bone turnover in healthy male adults. Calcif Tissue Int. 2003 Oct;73(4):356–360. doi: 10.1007/s00223-002-0018-9. [DOI] [PubMed] [Google Scholar]
- 105.Bergqvist AG, Schall JI, Stallings VA. Vitamin D status in children with intractable epilepsy, and impact of the ketogenic diet. Epilepsia. 2007 Jan;48(1):66–71. doi: 10.1111/j.1528-1167.2006.00803.x. [DOI] [PubMed] [Google Scholar]
- 106.Pack AM, Morrell MJ, Randall A, McMahon DJ, Shane E. Bone health in young women with epilepsy after one year of antiepileptic drug monotherapy. Neurology. 2008 Apr 29;70(18):1586–1593. doi: 10.1212/01.wnl.0000310981.44676.de. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kim SH, Lee JW, Choi KG, Chung HW, Lee HW. A 6-month longitudinal study of bone mineral density with antiepileptic drug monotherapy. Epilepsy Behav. 2007 Mar;10(2):291–295. doi: 10.1016/j.yebeh.2006.11.007. [DOI] [PubMed] [Google Scholar]
- 108.Cansu A, Yesilkaya E, Serdaroglu A, et al. Evaluation of bone turnover in epileptic children using oxcarbazepine. Pediatr Neurol. 2008 Oct;39(4):266–271. doi: 10.1016/j.pediatrneurol.2008.07.001. [DOI] [PubMed] [Google Scholar]
- 109.Hoikka V, Savolainen K, Alhava EM, Sivenius J, Karjalainen P, Parvianinen M. Anticonvulsant osteomalacia in epileptic outpatients. Ann Clin Res. 1982 Jun;14(3):129–132. [PubMed] [Google Scholar]
- 110.Rickers H, Deding A, Christiansen C, Rodbro P, Naestoft J. Corticosteroid-induced osteopenia and vitamin D metabolism. Effect of vitamin D2, calcium phosphate and sodium fluoride administration. Clin Endocrinol (Oxf) 1982 Apr;16(4):409–415. doi: 10.1111/j.1365-2265.1982.tb00734.x. [DOI] [PubMed] [Google Scholar]
- 111.Zerwekh JE, Emkey RD, Harris ED., Jr Low-dose prednisone therapy in rheumatoid arthritis: effect on vitamin D metabolism. Arthritis Rheum. 1984 Sep;27(9):1050–1052. doi: 10.1002/art.1780270913. [DOI] [PubMed] [Google Scholar]
- 112.Hahn TJ, Halstead LR, Baran DT. Effects of short term glucocorticoid administration on intestinal calcium absorption and circulating vitamin D metabolite concentrations in man. J Clin Endocrinol Metab. 1981 Jan;52(1):111–115. doi: 10.1210/jcem-52-1-111. [DOI] [PubMed] [Google Scholar]
- 113.Prummel MF, Wiersinga WM, Lips P, Sanders GT, Sauerwein HP. The course of biochemical parameters of bone turnover during treatment with corticosteroids. J Clin Endocrinol Metab. 1991 Feb;72(2):382–386. doi: 10.1210/jcem-72-2-382. [DOI] [PubMed] [Google Scholar]
- 114.Lems WF, Jacobs JW, Van Rijn HJ, Bijlsma JW. Changes in calcium and bone metabolism during treatment with low dose prednisone in young, healthy, male volunteers. Clin Rheumatol. 1995 Jul;14(4):420–424. doi: 10.1007/BF02207675. [DOI] [PubMed] [Google Scholar]
- 115.Halton JM, Atkinson SA, Fraher L, et al. Altered mineral metabolism and bone mass in children during treatment for acute lymphoblastic leukemia. J Bone Miner Res. 1996 Nov;11(11):1774–1783. doi: 10.1002/jbmr.5650111122. [DOI] [PubMed] [Google Scholar]
- 116.Hahn TJ, Halstead LR, Haddad JG., Jr Serum 25-hydroxyvitamin D concentrations in patients receiving chronic corticosteroid therapy. J Lab Clin Med. 1977 Aug;90(2):399–404. [PubMed] [Google Scholar]
- 117.Klein RG, Arnaud SB, Gallagher JC, Deluca HF, Riggs BL. Intestinal calcium absorption in exogenous hypercortisonism. Role of 25-hydroxyvitamin D and corticosteroid dose. J Clin Invest. 1977 Jul;60(1):253–259. doi: 10.1172/JCI108762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lund B, Storm TL, Melsen F, et al. Bone mineral loss, bone histomorphometry and vitamin D metabolism in patients with rheumatoid arthritis on long-term glucocorticoid treatment. Clin Rheumatol. 1985 Jun;4(2):143–149. doi: 10.1007/BF02032284. [DOI] [PubMed] [Google Scholar]
- 119.Bikle DD, Halloran B, Fong L, Steinbach L, Shellito J. Elevated 1,25-dihydroxyvitamin D levels in patients with chronic obstructive pulmonary disease treated with prednisone. J Clinical Endocrinol Metab. 1993 Feb;76(2):456–461. doi: 10.1210/jcem.76.2.8432789. [DOI] [PubMed] [Google Scholar]
- 120.Wetzsteon RJ, Shults J, Zemel BS, et al. Divergent effects of glucocorticoids on cortical and trabecular compartment BMD in childhood nephrotic syndrome. J Bone Miner Res. 2009 Mar;24(3):503–513. doi: 10.1359/JBMR.081101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wolthers OD, Riis BJ, Pedersen S. Bone turnover in asthmatic children treated with oral prednisolone or inhaled budesonide. Pediatr Pulmonol. 1993 Dec;16(6):341–346. doi: 10.1002/ppul.1950160604. [DOI] [PubMed] [Google Scholar]
- 122.Bijlsma JW, Duursma SA, Huber-Bruning O. Bone metabolism during methylprednisolone pulse therapy in rheumatoid arthritis. Ann Rheum Dis. 1986 Sep;45(9):757–760. doi: 10.1136/ard.45.9.757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Slovik DM, Neer RM, Ohman JL, et al. Parathyroid hormone and 25-hydroxyvitamin D levels in glucocorticoid-treated patients. Clin Endocrinol. 1980 Mar;12(3):243–248. doi: 10.1111/j.1365-2265.1980.tb02706.x. [DOI] [PubMed] [Google Scholar]
- 124.Als OS, Riis B, Christiansen C. Serum concentration of vitamin D metabolites in rheumatoid arthritis. Clin Rheumatol. 1987 Jun;6(2):238–243. doi: 10.1007/BF02201030. [DOI] [PubMed] [Google Scholar]
- 125.von Scheven E, Gordon CM, Wypij D, Wertz M, Gallagher KT, Bachrach L. Variable deficits of bone mineral despite chronic glucocorticoid therapy in pediatric patients with inflammatory diseases: a Glaser Pediatric Research Network study. J Pediatr Endocrinol Metab. 2006 Jun;19(6):821–830. doi: 10.1515/jpem.2006.19.6.821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Sentongo TA, Semaeo EJ, Stettler N, Piccoli DA, Stallings VA, Zemel BS. Vitamin D status in children, adolescents, and young adults with Crohn disease. Am J Clin Nutr. 2002 Nov;76(5):1077–1081. doi: 10.1093/ajcn/76.5.1077. [DOI] [PubMed] [Google Scholar]
- 127.Cohran VC, Griffiths M, Heubi JE. Bone mineral density in children exposed to chronic glucocorticoid therapy. Clin Pediatr. 2008;47(5):469–475. doi: 10.1177/0009922807311732. [DOI] [PubMed] [Google Scholar]
- 128.Santiago RA, Silva CA, Caparbo VF, Sallum AM, Pereira RM. Bone mineral apparent density in juvenile dermatomyositis: the role of lean body mass and glucocorticoid use. Scand J Rheumatol. 2008 Jan-Feb;37(1):40–47. doi: 10.1080/03009740701687226. [DOI] [PubMed] [Google Scholar]
- 129.Princen HM, Meijer P, Wolthers BG, Vonk RJ, Kuipers F. Cyclosporin A blocks bile acid synthesis in cultured hepatocytes by specific inhibition of chenodeoxycholic acid synthesis. Biochem J. 1991 Apr 15;275(Pt 2):501–505. doi: 10.1042/bj2750501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Winegar DA, Salisbury JA, Sundseth SS, Hawke RL. Effects of cyclosporin on cholesterol 27-hydroxylation and LDL receptor activity in HepG2 cells. J Lipid Res. 1996 Jan;37(1):179–191. [PubMed] [Google Scholar]
- 131.Souidi M, Parquet M, Ferezou J, Lutton C. Modulation of cholesterol 7alpha-hydroxylase and sterol 27-hydroxylase activities by steroids and physiological conditions in hamster. Life Sci. 1999;64(17):1585–1593. doi: 10.1016/s0024-3205(99)00089-2. [DOI] [PubMed] [Google Scholar]
- 132.Gueguen Y, Ferrari L, Souidi M, et al. Compared effect of immunosuppressive drugs cyclosporine A and rapamycin on cholesterol homeostasis key enzymes CYP27A1 and HMG-CoA reductase. Basic Clin Pharmacol Toxicol. 2007 Jun;100(6):392–397. doi: 10.1111/j.1742-7843.2007.00066.x. [DOI] [PubMed] [Google Scholar]
- 133.Grenet O, Bobadilla M, Chibout SD, Steiner S. Evidence for the impairment of the vitamin D activation pathway by cyclosporine A. Biochem Pharmacol. 2000 Feb 1;59(3):267–272. doi: 10.1016/s0006-2952(99)00321-4. [DOI] [PubMed] [Google Scholar]
- 134.Reichel H, Grussinger A, Knehans A, Kuhn K, Schmidt-Gayk H, Ritz E. Long-term therapy with cyclosporin A does not influence serum concentrations of vitamin D metabolites in patients with multiple sclerosis. Clin Investig. 1992 Jul;70(7):595–599. doi: 10.1007/BF00184801. [DOI] [PubMed] [Google Scholar]
- 135.de Sevaux RG, Hoitsma AJ, van Hoof HJ, Corstens FJ, Wetzels JF. Abnormal vitamin D metabolism and loss of bone mass after renal transplantation. Nephron Clin Pract. 2003 Jan;93(1):C21–28. doi: 10.1159/000066640. [DOI] [PubMed] [Google Scholar]
- 136.Saha HH, Salmela KT, Ahonen PJ, et al. Sequential changes in vitamin D and calcium metabolism after successful renal transplantation. Scand J Urol Nephrol. 1994 Mar;28(1):21–27. doi: 10.3109/00365599409180465. [DOI] [PubMed] [Google Scholar]
- 137.Shaw AJ, Hayes ME, Davies M, et al. Cyclosporin A and vitamin D metabolism: studies in patients with psoriasis and in rats. Clin Sci. 1994 May;86(5):627–632. doi: 10.1042/cs0860627. [DOI] [PubMed] [Google Scholar]
- 138.Edwards BD, Davies M, Mawer EB, Chalmers RJ, Testa HJ, Ballardie FW. Interrelationship between serum concentrations of 1,25-dihydroxyvitamin D, parathyroid hormone and renal haemodynamics after low dose cyclosporin. Miner Electrolyte Metab. 1994;20(5):250–254. [PubMed] [Google Scholar]
- 139.Falkiewicz K, Kaminska D, Nahaczewska W, et al. Renal function and tubular phosphate handling in long-term cyclosporine- and tacrolimus-based immunosuppression in kidney transplantation. Transplant Proc. 2006 Jan-Feb;38(1):119–122. doi: 10.1016/j.transproceed.2005.12.083. [DOI] [PubMed] [Google Scholar]
- 140.Iwasaki K. Metabolism of tacrolimus (FK506) and recent topics in clinical pharmacokinetics. Drug Metab Pharmacokinet. 2007 Oct;22(5):328–335. doi: 10.2133/dmpk.22.328. [DOI] [PubMed] [Google Scholar]
- 141.Meijerman I, Beijnen JH, Schellens JH. Herb-drug interactions in oncology: focus on mechanisms of induction. Oncologist. 2006 Jul-Aug;11(7):742–752. doi: 10.1634/theoncologist.11-7-742. [DOI] [PubMed] [Google Scholar]
- 142.Kailajarvi ME, Salminen EK, Paija OM, Virtanent AM, Leino AE, Irjala KA. Serum bone markers in breast cancer patients during 5-fluorouracil, epirubicin and cyclophosphamide (FEC) therapy. Anticancer Res. 2004 Mar-Apr;24(2C):1271–1274. [PubMed] [Google Scholar]
- 143.Gao Y, Shimizu M, Yamada S, Ozaki Y, Aso T. The effects of chemotherapy including cisplatin on vitamin D metabolism. Endocr J. 1993 Dec;40(6):737–742. doi: 10.1507/endocrj.40.737. [DOI] [PubMed] [Google Scholar]
- 144.Fakih MG, Trump DL, Johnson CS, Tian L, Muindi J, Sunga AY. Chemotherapy is linked to severe vitamin D deficiency in patients with colorectal cancer. Int J Colorectal Dis. 2009 Feb;24(2):219–224. doi: 10.1007/s00384-008-0593-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Flexner C. Antiretroviral agents and treatment of HIV infection. In: Brunton LL, Lazo JS, Parker KL, editors. Goodman & Gilman’s The Pharmacological Basis of Therapeutics. 11. New York: McGraw-Hill; 2006. [Google Scholar]
- 146.Zhou SF, Xue CC, Yu XQ, Li C, Wang G. Clinically important drug interactions potentially involving mechanism-based inhibition of cytochrome P450 3A4 and the role of therapeutic drug monitoring. Ther Drug Monit. 2007 Dec;29(6):687–710. doi: 10.1097/FTD.0b013e31815c16f5. [DOI] [PubMed] [Google Scholar]
- 147.Cozzolino M, Vidal M, Arcidiacono MV, Tebas P, Yarasheski KE, Dusso AS. HIV-protease inhibitors impair vitamin D bioactivation to 1,25-dihydroxyvitamin D. Aids. 2003 Mar 7;17(4):513–520. doi: 10.1097/00002030-200303070-00006. [DOI] [PubMed] [Google Scholar]
- 148.Ramayo E, Gonzalez-Moreno MP, Macias J, et al. Relationship between osteopenia, free testosterone, and vitamin D metabolite levels in HIV-infected patients with and without highly active antiretroviral therapy. AIDS Res Hum Retroviruses. 2005 Nov;21(11):915–921. doi: 10.1089/aid.2005.21.915. [DOI] [PubMed] [Google Scholar]
- 149.Curtis JR, Smith B, Weaver M, et al. Ethnic variations in the prevalence of metabolic bone disease among HIV-positive patients with lipodystrophy. AIDS Res Hum Retroviruses. 2006 Feb;22(2):125–131. doi: 10.1089/aid.2006.22.125. [DOI] [PubMed] [Google Scholar]
- 150.Garcia Aparicio AM, Munoz Fernandez S, Gonzalez J, et al. Abnormalities in the bone mineral metabolism in HIV-infected patients. Clin Rheumatol. 2006 Jul;25(4):537–539. doi: 10.1007/s10067-005-0028-x. [DOI] [PubMed] [Google Scholar]
- 151.Bengoa JM, Bolt MJ, Rosenberg IH. Hepatic vitamin D 25-hydroxylase inhibition by cimetidine and isoniazid. J Lab Clin Med. 1984 Oct;104(4):546–552. [PubMed] [Google Scholar]
- 152.Wyatt CL, Jensen LS, Rowland GN., 3rd Effect of cimetidine on eggshell quality and plasma 25-hydroxycholecalciferol in laying hens. Poult Sci. 1990 Nov;69(11):1892–1899. doi: 10.3382/ps.0691892. [DOI] [PubMed] [Google Scholar]
- 153.Odes HS, Fraser GM, Krugliak P, Lamprecht SA, Shany S. Effect of cimetidine on hepatic vitamin D metabolism in humans. Digestion. 1990;46(2):61–64. doi: 10.1159/000200333. [DOI] [PubMed] [Google Scholar]
- 154.Richards DA. Comparative pharmacodynamics and pharmacokinetics of cimetidine and ranitidine. J Clin Gastroenterol. 1983;5(Suppl 1):81–90. doi: 10.1097/00004836-198312001-00008. [DOI] [PubMed] [Google Scholar]
- 155.Hathcock JN, Shao A, Vieth R, Heaney R. Risk assessment for vitamin D. Am J Clin Nutr. 2007 Jan;85(1):6–18. doi: 10.1093/ajcn/85.1.6. [DOI] [PubMed] [Google Scholar]
- 156.Parfitt AM. Chlorothiazide-induced hypercalcemia in juvenile osteoporosis and hyperparathyroidism. N Engl J Med. 1969 Jul 10;281(2):55–59. doi: 10.1056/NEJM196907102810201. [DOI] [PubMed] [Google Scholar]
- 157.Lichtwitz A, Parlier R, de S, Hioco D, Miravet L. The hypocalciuric effect of diuretic sulfonamides. Sem Med Prof Med Soc. 1961 Aug 14–20;37:2350–2362. [PubMed] [Google Scholar]
- 158.Torsti P, Lamberg BA. The effect of a two-day treatment with chlorothiazide on the urinary excretion of calcium, phosphate and sodium in hyper- and hypocalcemia. Acta Med Scand. 1964;175(Suppl 412):181–191. doi: 10.1111/j.0954-6820.1964.tb04649.x. [DOI] [PubMed] [Google Scholar]
- 159.Higgins BA, Nassim JR, Collins J, Hilb A. The effect of bendrofluazide on urine calcium exretion. Clin Sci. 1964 Dec;27:457–462. [PubMed] [Google Scholar]
- 160.Drinka PJ, Nolten WE. Hazards of treating osteoporosis and hypertension concurrently with calcium, vitamin D, and distal diuretics. J Am Geriatr Soc. 1984 May;32(5):405–407. doi: 10.1111/j.1532-5415.1984.tb02050.x. [DOI] [PubMed] [Google Scholar]
- 161.Crowe M, Wollner L, Griffiths RA. Hypercalcaemia following vitamin D and thiazide therapy in the elderly. Practitioner. 1984 Mar;228(1389):312–313. [PubMed] [Google Scholar]
- 162.Riis B, Christiansen C. Actions of thiazide on vitamin D metabolism: a controlled therapeutic trial in normal women early in the postmenopause. Metab Clin Exp. 1985 May;34(5):421–424. doi: 10.1016/0026-0495(85)90206-9. [DOI] [PubMed] [Google Scholar]
- 163.Lemann J, Jr, Gray RW, Maierhofer WJ, Cheung HS. Hydrochlorothiazide inhibits bone resorption in men despite experimentally elevated serum 1,25-dihydroxyvitamin D concentrations. Kidney Int. 1985 Dec;28(6):951–958. doi: 10.1038/ki.1985.223. [DOI] [PubMed] [Google Scholar]
- 164.Kokot F, Pietrek J, Srokowska S, et al. 25-hydroxyvitamin D in patients with essential hypertension. Clin Nephrol. 1981 Oct;16(4):188–192. [PubMed] [Google Scholar]
- 165.Perry HM, 3rd, Jensen J, Kaiser FE, Horowitz M, Perry HM, Jr, Morley JE. The effects of thiazide diuretics on calcium metabolism in the aged. J Am Geriatr Soc. 1993 Aug;41(8):818–822. doi: 10.1111/j.1532-5415.1993.tb06176.x. [DOI] [PubMed] [Google Scholar]
- 166.Huang H, Wang H, Sinz M, et al. Inhibition of drug metabolism by blocking the activation of nuclear receptors by ketoconazole. Oncogene. 2007 Jan 11;26(2):258–268. doi: 10.1038/sj.onc.1209788. [DOI] [PubMed] [Google Scholar]
- 167.Marechal JD, Yu J, Brown S, et al. In silico and in vitro screening for inhibition of cytochrome P450 CYP3A4 by comedications commonly used by patients with cancer. Drug Metab Dispos. 2006 Apr;34(4):534–538. doi: 10.1124/dmd.105.007625. [DOI] [PubMed] [Google Scholar]
- 168.Loose DS, Kan PB, Hirst MA, Marcus RA, Feldman D. Ketoconazole blocks adrenal steroidogenesis by inhibiting cytochrome P450-dependent enzymes. J Clin Invest. 1983 May;71(5):1495–1499. doi: 10.1172/JCI110903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Hollis BW, Horst RL. The assessment of circulating 25(OH)D and 1,25(OH)2D: where we are and where we are going. J Steroid Biochem Mol Biol. 2007 Mar;103(3–5):473–476. doi: 10.1016/j.jsbmb.2006.11.00. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Binkley N, Krueger D, Cowgill CS, et al. Assay variation confounds the diagnosis of hypovitaminosis D: a call for standardization. J Clin Endocrinol Metab. 2004 Jul;89(7):3152–3157. doi: 10.1210/jc.2003-031979. [DOI] [PubMed] [Google Scholar]
- 171.Carter GD, Carter CR, Gunter E, et al. Measurement of Vitamin D metabolites: an international perspective on methodology and clinical interpretation. J Steroid Biochem Mol Biol. 2004 May;89–90(1–5):467–471. doi: 10.1016/j.jsbmb.2004.03.055. [DOI] [PubMed] [Google Scholar]
- 172.Tai SS, Bedner M, Phinney KW. Development of a candidate reference measurement procedure for the determination of 25-hydroxyvitamin D3 and 25-hydroxyvitamin D2 in human serum using isotope-dilution liquid chromatography-tandem mass spectrometry. Anal Chem. Mar 1;82(5):1942–1948. doi: 10.1021/ac9026862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002 Feb-May;34(1–2):83–448. doi: 10.1081/dmr-120001392. [DOI] [PubMed] [Google Scholar]
- 174.Zhou S, Yung Chan S, Cher Goh B, et al. Mechanism-based inhibition of cytochrome P450 3A4 by therapeutic drugs. Clin Pharmacokinet. 2005;44(3):279–304. doi: 10.2165/00003088-200544030-00005. [DOI] [PubMed] [Google Scholar]
- 175.Sjostrom L, Rissanen A, Andersen T, et al. Randomised placebo-controlled trial of orlistat for weight loss and prevention of weight regain in obese patients. European Multicentre Orlistat Study Group. Lancet. 1998 Jul 18;352(9123):167–172. doi: 10.1016/s0140-6736(97)11509-4. [DOI] [PubMed] [Google Scholar]
- 176.Pack AM, Morrell MJ, Marcus R, et al. Bone mass and turnover in women with epilepsy on antiepileptic drug monotherapy. Ann Neurol. 2005;57(2):252–257. doi: 10.1002/ana.20378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Searing DA, Zhang Y, Murphy JR, Hauk PJ, Goleva E, Leung DY. Decreased serum vitamin D levels in children with asthma are associated with increased corticosteroid use. J Allergy Clin Immunol. 2010 May;125(5):995–1000. doi: 10.1016/j.jaci.2010.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Sutherland ER, Goleva E, Jackson LP, Stevens AD, Leung DY. Vitamin D levels, lung function, and steroid response in adult asthma. Am J Respir Crit Care Med. 2010 Apr 1;181(7):699–704. doi: 10.1164/rccm.200911-1710OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Toloza SMA, Cole DEC, Gladman DD, Ibanez D, Urowitz MB. Vitamin D insufficiency in a large female SLE cohort. Lupus. 2010;19:13–19. doi: 10.1177/0961203309345775. [DOI] [PubMed] [Google Scholar]
- 180.Nordal KP, Dahl E, Halse J, Aksnes L, Thomassen Y, Flatmark A. Aluminum metabolism and bone histology after kidney transplantation: a one-year follow-up study. J Clin Endocrinol Metab. 1992 May;74(5):1140–1145. doi: 10.1210/jcem.74.5.1569161. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.