Methods and compositions for reducing parathyroid levels

Information

  • Patent Grant
  • 11672809
  • Patent Number
    11,672,809
  • Date Filed
    Tuesday, March 29, 2011
    13 years ago
  • Date Issued
    Tuesday, June 13, 2023
    11 months ago
Abstract
Methods and compositions for reducing serum parathyroid levels in, for example, chronic kidney disease patients are disclosed. In these methods, an effective amount of a modified release formulation of 25-hydroxy vitamin D is orally administered to a patient suffering from secondary hyperparathyroidism to lower the patient's serum intact parathyroid hormone (iPTH) level, while avoiding a surge in serum total 25-hydroxy vitamin D.
Description
BACKGROUND
Field of the Disclosure

The disclosure relates generally to methods and compositions for reducing serum parathyroid levels, for example in chronic kidney disease patients. More particularly, the invention relates to methods and compositions including controlled-release formulations of 25-hydroxyvitamin D for oral delivery.


Brief Description of Related Technology

Cholecalciferol and ergocalciferol (collectively are referred to as “Vitamin D”) are fat-soluble seco-steroid precursors to Vitamin D prohormones. The Vitamin D metabolites known as 25-hydroxyvitamin D2 and 25-hydroxyvitamin D3 (collectively referred to herein as “25-hydroxyvitamin D”) are fat-soluble steroid prohormones to Vitamin D hormones that contribute to the maintenance of normal levels of calcium and phosphorus in the bloodstream.


Cholecalciferol and ergocalciferol are normally present at stable, low concentrations in human blood. Slight, if any increases in blood Vitamin D levels occur after meals since unsupplemented diets have low Vitamin D content, even those containing foods fortified with Vitamin D. Almost all human Vitamin D supply comes from fortified foods, exposure to sunlight or from dietary supplements, with the latter source becoming increasingly important. Blood Vitamin D levels rise only gradually, if at all, after sunlight exposure since cutaneous 7-dehydroxycholesterol is modified by ultraviolet (UV) radiation to pre-Vitamin D3, which undergoes thermal conversion in the skin to cholecalciferol over a period of several days before circulating in the blood. In contrast, supplements such as those currently available, do cause rapid and marked increases in intraluminal, blood and intracellular levels of Vitamin D proportional to the dose administered.


Both cholecalciferol and ergocalciferol are metabolized into prohormones by enzymes primarily located in the liver of the human body. Cholecalciferol is metabolized into a prohormone 25-hydroxyvitamin D3, and ergocalciferol is metabolized into two prohormones, 25-hydroxyvitamin D2 and 24(S)-hydroxyvitamin D2. Cholecalciferol and ergocalciferol also can be metabolized into prohormones outside of the liver in certain cells, such as enterocytes, by enzymes which are identical or similar to those found in the liver. Elevating concentrations of either precursor increases prohormone production; similarly, lowering precursor concentrations decreases hormone production. Surges in the blood levels of cholecalciferol and/or ergocalciferol (“cholecalciferol/ergocalciferol”) can transiently raise intracellular Vitamin D concentrations, accelerating prohormone production and elevating intracellular and blood prohormone concentrations. Surges in the blood levels of cholecalciferol and/or ergocalciferol also can saturate the enzymes which produce the prohormones, causing the excess Vitamin D to be catabolized or shunted to long-term storage in adipose tissue. Vitamin D stored in adipose tissue is less available for future conversion to prohormones, due to local catabolism. Surges in intraluminal levels of Vitamin D after ingestion of current oral supplements can directly boost Vitamin D and prohormone concentrations in the local enterocytes, thereby exerting “first pass” effects on calcium and phosphorus metabolism in the small intestine.


The Vitamin D prohormones are further metabolized in the kidneys into potent hormones. The prohormone 25-hydroxyvitamin D3 is metabolized into a hormone 1α,25-dihydroxyvitamin D3 (or calcitriol); likewise, 25-hydroxyvitamin D2 and 24(S)-hydroxyvitamin D2 are metabolized into hormones known as 1α,25-dihydroxyvitamin D2 and 1α,24(S)-dihydroxyvitamin D2, respectively. Production of these hormones from the prohormones also can occur outside of the kidney in cells which contain the required enzyme(s).


Surges in blood or intracellular prohormone concentrations can promote excessive extrarenal hormone production, leading to local adverse effects on calcium and phosphorus metabolism. Such surges also can inhibit hepatic prohormone production from subsequent supplemental Vitamin D and promote catabolism of both Vitamin D and 25-hydroxyvitamin D in the kidney and other tissues.


Blood Vitamin D hormone concentrations remain generally constant through the day in healthy individuals, but can vary significantly over longer periods of time in response to seasonal changes in sunlight exposure or sustained changes in Vitamin D intake. Normally, blood levels of cholecalciferol, ergocalciferol and the three Vitamin D prohormones are also constant through the day, given a sustained, adequate supply of Vitamin D from sunlight exposure and an unsupplemented diet. Blood levels of cholecalciferol and ergocalciferol, however, can increase markedly after administration of currently available Vitamin D supplements, especially at doses which greatly exceed the amounts needed to prevent Vitamin D deficiency, rickets or osteomalacia.


The Vitamin D hormones have essential roles in human health which are mediated by intracellular Vitamin D receptors (VDR). In particular, the Vitamin D hormones regulate blood calcium levels by controlling the absorption of dietary calcium by the small intestine and the reabsorption of calcium by the kidneys. Excessive hormone levels can lead to abnormally elevated urine calcium (hypercalciuria), blood calcium (hypercalcemia) and blood phosphorus (hyperphosphatemia). The Vitamin D hormones also participate in the regulation of cellular differentiation and growth, parathyroid hormone (PTH) secretion by the parathyroid glands, and normal bone formation and metabolism. Further, Vitamin D hormones are required for the normal functioning of the musculoskeletal, immune and renin-angiotensin systems. Numerous other roles for Vitamin D hormones are being postulated and elucidated based on the documented presence of intracellular VDR in nearly every human tissue.


Secondary hyperparathyroidism is a disorder which develops primarily because of Vitamin D insufficiency. It is characterized by abnormally elevated blood levels of PTH and, in the absence of early detection and treatment, it becomes associated with parathyroid gland hyperplasia and a constellation of metabolic bone diseases. It is a common complication of chronic kidney disease (CKD), with rising incidence as CKD progresses. Secondary hyperparathyroidism can also develop in individuals with healthy kidneys, due to environmental, cultural or dietary factors which prevent adequate Vitamin D supply.


As to secondary hyperparathyroidism and its occurrence in CKD, there is a progressive loss of cells of the proximal nephrons, the primary site for the synthesis of the vitamin D hormones (collectively “1,25-dihydroxyvitamin D”) from 25-hydroxyvitamin D3 and 25-hydroxyvitamin D2. In addition, the loss of functioning nephrons leads to retention of excess phosphorus which reduces the activity of the renal 25-hydroxyvitamin D-1α-hydroxylase, the enzyme which catalyzes the reaction to produce the D hormones. These two events account for the low serum levels of 1,25-dihydroxyvitamin D commonly found in patients with moderate to severe CKD when Vitamin D supply is adequate.


Reduced serum levels of 1,25-dihydroxyvitamin D cause increased, and ultimately excessive, secretion of PTH by direct and indirect mechanisms. The resulting hyperparathyroidism leads to markedly increased bone turnover and its sequela of renal osteodystrophy, which may include a variety of other diseases, such as, osteitis fibrosa cystica, osteomalacia, osteoporosis, extraskeletal calcification and related disorders, e.g., bone pain, periarticular inflammation and Mockerberg's sclerosis. Reduced serum levels of 1,25-dihydroxyvitamin D also can cause muscle weakness and growth retardation with skeletal deformities (most often seen in pediatric patients).


Blood levels of 1,25-dihydroxyvitamin D are precisely regulated by a feedback mechanism which involves PTH and vitamin D hormone. The renal 1α-hydroxylase (or CYP27B1) is stimulated by PTH and inhibited by 1,25-dihydroxyvitamin D. When blood levels of 1,25-dihydroxyvitamin D fall, the parathyroid glands sense this change via intracellular Vitamin D receptors and secrete PTH. The secreted PTH stimulates expression of renal CYP27B1 and, thereby, increases production of Vitamin D hormones. As blood concentrations of 1,25-dihydroxyvitamin D rise again, the parathyroid glands attenuate further PTH secretion. As blood PTH levels fall, renal production of Vitamin D hormones decreases. Rising blood levels of 1,25-dihydroxyvitamin D also directly inhibit further Vitamin D hormone production by CYP27B1.


PTH secretion can be abnormally and excessively suppressed in situations where blood 1,25-dihydroxyvitamin D concentrations become excessively elevated, as can occur in certain disorders such as sarcoidosis or as a result of bolus doses of Vitamin D hormone replacement therapies. Oversuppression of PTH secretion can cause or exacerbate disturbances in calcium homeostasis. The parathyroid glands and the renal CYP27B1 are exquisitely sensitive to changes in blood concentrations of Vitamin D hormones such that serum 1,25-dihydroxyvitamin D is tightly controlled, fluctuating up or down by less than 20% during any 24-hour period. In contrast to renal production of Vitamin D hormones, extrarenal production is not under precise feedback control.


Blood levels of 1,25-dihydroxyvitamin D and substrate 25-hydroxyvitamin D prohormone, and regulation thereof, can also be affected by vitamin D hormone analogs, such as 1α-hydroxyvitamin D2 and 19-nor-1,25 dihydroxyvitamin D2.


The actions of Vitamin D hormones on specific tissues depend on the degree to which they bind to (or occupy) the intracellular VDR in those tissues. Cholecalciferol and ergocalciferol have affinities for the VDR which are estimated to be at least 100-fold lower than those of the Vitamin D hormones. As a consequence, physiological concentrations of cholecalciferol and ergocalciferol exert little, if any, biological actions without prior metabolism to Vitamin D hormones. However, supraphysiologic levels of cholecalciferol and ergocalciferol, in the range of 10 to 1,000 fold higher than normal, can sufficiently occupy the VDR and exert actions like the Vitamin D hormones. Similarly, the prohormones 25-hydroxyvitamin D2 and 25-hydroxyvitamin D3 have essentially identical affinities for the VDR which are also estimated to be at least 100-fold lower than those of the Vitamin D hormones. As a consequence, physiological concentrations of 25-hydroxyvitamin D2 and 25-hydroxyvitamin D3 have little, if any, biological actions without prior metabolism to Vitamin D hormones. However, supraphysiologic levels of 25-hydroxyvitamin D2 and 25-hydroxyvitamin D3, in the range of 10 to 1,000 fold higher than normal, can sufficiently occupy the VDR to exert actions like the Vitamin D hormones.


Production of Vitamin D prohormones declines when Vitamin D is in short supply, as in conditions such as Vitamin D insufficiency or Vitamin D deficiency (alternatively, hypovitaminosis D). Low production of Vitamin D prohormones leads to low blood levels of 25-hydroxyvitamin D. Inadequate Vitamin D supply often develops in individuals who are infrequently exposed to sunlight, have chronically inadequate intakes of Vitamin D, or suffer from conditions that reduce the intestinal absorption of fat soluble vitamins (such as Vitamin D). It has recently been reported that most individuals living in northern latitudes have inadequate Vitamin D supplies. Left untreated, inadequate Vitamin D supply can cause serious bone disorders, including rickets and osteomalacia.


The Institute of Medicine (IOM) of the National Academy of Sciences has concluded that an Adequate Intake (AI) of Vitamin D for a healthy individual ranges from 200 to 600 IU per day, depending on the individual's age and sex (see Standing Committee on the Scientific Evaluation of Dietary Reference Intakes, Dietary reference intakes: calcium, phosphorus, magnesium, vitamin D, and fluoride, Washington, D.C.: National Academy Press 1997, incorporated herein by reference). The AI for Vitamin D was defined primarily on the basis of a serum 25-hydroxyvitamin D level sufficient to prevent Vitamin D deficiency, rickets or osteomalacia (or at least 11 ng/mL). The IOM also established a Tolerable Upper Intake Level (UL) for Vitamin D of 2,000 IU per day, based on evidence that higher doses are associated with an increased risk of hypercalciuria, hypercalcemia and related sequelae, including cardiac arrhythmias, seizures, and generalized vascular and other soft-tissue calcification.


Currently available oral Vitamin D supplements are far from ideal for achieving and maintaining optimal blood 25-hydroxyvitamin D levels. These preparations typically contain 400 IU to 5,000 IU of Vitamin D3 or 50,000 IU of Vitamin D2 and are formulated for quick or immediate release in the gastrointestinal tract. When administered at chronically high doses, as is often required for Vitamin D repletion, these products have significant and, often, severe limitations which are summarized below.


High doses of immediate release Vitamin D supplements produce marked surges in blood Vitamin D levels, thereby promoting: (a) storage of Vitamin D in adipose tissue, which is undesirable because stored Vitamin D is less available for later hepatic conversion to 25-hydroxyvitamin D; (b) hepatic catabolism of Vitamin D to metabolites, which are less useful or no longer useful for boosting blood 25-hydroxyvitamin D levels, via 24- and/or 26-hydroxylation; and, (c) excessive intracellular 24- or 25-hydroxylation of Vitamin D, which leads to increased risk of hypercalciuria, hypercalcemia and hyperphosphatemia.


High doses of immediate release Vitamin D supplements also produce surges or spikes in blood and intracellular 25-hydroxyvitamin D levels, thereby promoting: (a) excessive extrarenal production of Vitamin D hormones, and leading to local aberrations in calcium and phosphorus homeostasis and increased risk of hypercalciuria, hypercalcemia and hyperphosphatemia; (b) accelerated catabolism of both Vitamin D and 25-hydroxyvitamin D by 24- and/or 26-hydroxylation in the kidney and other tissues; (c) down-regulation of hepatic production of Vitamin D prohormones, unnecessarily impeding the efficient repletion of Vitamin D insufficiency or deficiency; and, (d) local aberrations in calcium and phosphorus homeostasis mediated by direct binding to VDR.


Furthermore, high doses of immediate release Vitamin D supplements produce supraphysiologic pharmacological concentrations of Vitamin D, e.g., in the lumen of the duodenum, promoting: (a) 25-hydroxylation in the enterocytes and local stimulation of intestinal absorption of calcium and phosphorus, leading to increased risk of hypercalciuria, hypercalcemia and hyperphosphatemia; (b) catabolism of Vitamin D by 24- and/or 26-hydroxylation in the local enterocytes, causing decreased systemic bioavailability; and (c) absorption primarily via chylomicrons, leading to increased hepatic catabolism.


Vitamin D supplementation above the UL is frequently needed in certain individuals; however, currently available oral Vitamin D supplements are not well suited for maintaining blood 25-hydroxyvitamin D levels at optimal levels given the problems of administering high doses of immediate release Vitamin D compounds.


Blood Vitamin D hormone concentrations also remain generally constant through the day in healthy individuals, but can vary significantly over longer periods of time in response to seasonal changes in sunlight exposure or sustained alterations in Vitamin D intake. Marked differences in normal Vitamin D hormone levels are commonly observed among healthy individuals, with some individuals having stable concentrations as low as approximately 20 pg/mL and others as high as approximately 70 pg/mL. Due to this wide normal range, medical professionals have difficulty interpreting isolated laboratory determinations of serum total 1,25-dihydroxyvitamin D; a value of 25 pg/mL may represent a normal value for one individual or a relative deficiency in another.


Transiently low blood levels of 1,25-dihydroxyvitamin D stimulate the parathyroid glands to secrete PTH for brief periods ending when normal blood Vitamin D hormone levels are restored. In contrast, chronically low blood levels of 1,25-dihydroxyvitamin D continuously stimulate the parathyroid glands to secrete PTH, resulting in a disorder known as secondary hyperparathyroidism. Chronically low hormone levels also decrease intestinal calcium absorption, leading to reduced blood calcium concentrations (hypocalcemia) which further stimulate PTH secretion. Continuously stimulated parathyroid glands become increasingly hyperplastic and eventually develop resistance to regulation by vitamin D hormones. Without early detection and treatment, secondary hyperparathyroidism progressively increases in severity, causing debilitating metabolic bone diseases, including osteoporosis and renal osteodystrophy.


Chronically low blood levels of 1,25-dihydroxyvitamin D develop when there is insufficient renal CYP27B1 to produce the required supply of Vitamin D hormones, a situation which commonly arises in CKD. The activity of renal CYP27B1 declines as the Glomerular Filtration Rate (GFR) falls below approximately 60 ml/min/1.73 m2 due to the loss of functioning nephrons. In end-stage renal disease (ESRD), when the kidneys fail completely and hemodialysis is required for survival, renal CYP27B1 often becomes altogether absent. Any remaining CYP27B1 is greatly inhibited by elevated serum phosphorous (hyperphosphatemia) caused by inadequate renal excretion of dietary phosphorous.


Chronically low blood levels of 1,25-dihydroxyvitamin D also develop because of an insufficiency of Vitamin D prohormones, since renal hormone production cannot proceed without the required precursors. Prohormone production declines markedly when cholecalciferol and ergocalciferol are in short supply, a condition often described by terms such as “Vitamin D insufficiency,” “Vitamin D deficiency,” or “hypovitaminosis D.” Therefore, measurement of 25-hydroxyvitamin D levels in blood has become the accepted method among healthcare professionals to monitor Vitamin D status. Recent studies have documented that the great majority of CKD patients have low blood levels of 25-hydroxyvitamin D, and that the prevalence of Vitamin D insufficiency and deficiency increases as CKD progresses.


It follows that individuals most vulnerable to developing chronically low blood levels of 1,25-dihydroxyvitamin D are those with CKD. Most CKD patients typically have decreased levels of renal CYP27B1 and a shortage of 25-hydroxyvitamin D prohormones. Not surprisingly, most CKD patients develop secondary hyperparathyroidism. Unfortunately, early detection and treatment of secondary hyperparathyroidism in CKD is rare, let alone prevention.


The National Kidney Foundation (NKF) has focused the medical community's attention on the need for early detection and treatment of secondary hyperparathyroidism by publishing Kidney Disease Outcomes Quality Initiative (K/DOQI) Clinical Practice Guidelines for Bone Metabolism and Disease in Chronic Kidney Disease [Am. J. Kidney Dis. 42:S1-S202, 2003] and, subsequently, the Kidney Disease Improving Global Outcomes (KDIGO) Clinical Practice Guidelines for Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD) [Kidney International Suppl. 113:S1-130, 2009]. Of the two Guidelines, the former offered the most detailed guidance to physicians. The K/DOQI Guidelines identified the primary etiology of secondary hyperparathyroidism as chronically low blood levels of 1,25-dihydroxyvitamin D and recommended regular screening in CKD Stages 3 through 5 for elevated blood PTH levels relative to stage-specific PTH target ranges, which for Stage 3 is 35-70 pg/mL (equivalent to 3.85-7.7 pmol/L), for Stage 4 is 70-110 pg/mL (equivalent to 7.7-12.1 pmol/L), and for Stage 5 is 150-300 pg/mL (equivalent to 16.5-33.0 pmol/L) (defined in K/DOQI Guideline No. 1). In the event that screening revealed an iPTH value to be above the ranges targeted for CKD Stages 3 and 4, the Guidelines recommended a follow-up evaluation of serum total 25-hydroxyvitamin D to detect possible Vitamin D insufficiency or deficiency. If 25-hydroxyvitamin D below 30 ng/mL was observed, the recommended intervention was Vitamin D repletion therapy using orally administered ergocalciferol. If 25-hydroxyvitamin D above 30 ng/mL was observed, the recommended intervention was Vitamin D hormone replacement therapy using known oral or intravenous Vitamin D hormones or analogs. The Guidelines did not recommend the concurrent application of Vitamin D repletion and Vitamin D hormone replacement therapies, consistent with warnings mandated by the Food and Drug Administration in package inserts for Vitamin D hormone replacement products.


The NKF K/DOQI Guidelines defined Vitamin D sufficiency as serum 25-hydroxyvitamin D levels ≥30 ng/mL. Recommended Vitamin D repletion therapy for patients with “Vitamin D insufficiency,” defined as serum total 25-hydroxyvitamin D of 16-30 ng/mL, was 50,000 IU per month of oral Vitamin D2 for 6 months, given either in single monthly doses or in divided doses of approximately 1,600 IU per day. Recommended repletion therapy for patients with “Vitamin D deficiency” was more aggressive: for “mild” deficiency, defined as serum total 25-hydroxyvitamin D of 5-15 ng/mL, the Guidelines recommended 50,000 IU per week of oral Vitamin D2 for 4 weeks, followed by 50,000 IU per month for another 5 months; for “severe” deficiency, defined as serum 25-hydroxyvitamin D below 5 ng/mL, the Guidelines recommended 50,000 IU/week of oral Vitamin D2 for 12 weeks, followed by 50,000 IU/month for another 3 months. Doses of 50,000 IU per week are approximately equivalent to 7,000 IU per day.


SUMMARY

A first aspect of the disclosure is a method of treating secondary hyperparathyroidism, including orally administering to a human patient suffering from secondary hyperparathyroidism an effective amount of a modified release dosage form including 25-hydroxyvitamin D as described herein to provide a rise in serum total 25-hydroxyvitamin D of at least 7 ng/ml and no greater than 30 ng/ml within the first 24 hours after the administering and lower the patient's serum intact parathyroid hormone (iPTH) level.


Optionally, the amount of the modified release dosage form administered is effective to provide a rise in serum total 25-hydroxyvitamin D of at least 8 ng/ml and no greater than 16 ng/ml within the first 24 hours after dosing. Optionally, the amount of the modified release dosage form administered is effective to provide a rise in serum total 25-hydroxyvitamin D of at least 10 ng/ml and no greater than 14 ng/ml within the first 24 hours after dosing.


In any one of the foregoing methods, optionally the patient's serum total 25-hydroxyvitamin D can be raised to at least 30 ng/ml.


In any one of the foregoing methods, optionally the patient's iPTH level can be lowered by at least 10% compared to baseline, or at least 15% compared to baseline.


In any one of the foregoing methods, optionally the patient's iPTH level remains lowered for at least 48 hours.


In any one of the foregoing methods, optionally the patient's iPTH level remains lowered for at least 7 days.


In any one of the foregoing methods, optionally the method of treatment further includes performing the administering step on a frequency of every other day, or less. In any one of the foregoing methods, optionally the frequency is weekly. In any one of the foregoing methods, optionally the frequency is monthly.


In any one of the foregoing methods, optionally the bioavailable amount of the 25-hydroxyvitamin D administered is greater than 45 μg, optionally in a range of 70 to 110 μg.


In any one of the foregoing methods, optionally the patient suffers from hyperparathyroidism secondary to chronic kidney disease (CKD). In any one of the foregoing methods, optionally the CKD is Stage 2, Stage 3, Stage 4, or Stage 5 CKD. In any one of the foregoing methods, optionally the CDK is Stage 3 or Stage 4.


In any one of the foregoing methods, optionally the amount of the modified release dosage form administered can be further effective to provide a rise in serum total 1,25-dihydroxyvitamin D of greater than 3 pg/ml and further less than 10 pg/ml within the first 24 hours after dosing. In any one of the foregoing methods, optionally the rise in serum total 1,25-dihydroxyvitamin D can be at least 7 pg/ml and further can be less than 10 pg/ml within the first 48 hours after dosing.


In any one of the foregoing methods, optionally the Tmax of serum total 25-hydroxyvitamin D following the administering step can be at least 4 hours. In any one of the foregoing methods, optionally the Tmax of serum total 25-hydroxyvitamin D following the administering step can be at least 12 hours.


In any one of the foregoing methods, optionally the patient can be one who also suffers from vitamin D insufficiency or deficiency, as defined by serum total 25-hydroxyvitamin D of less than 30 ng/ml.


In any one of the foregoing methods, optionally the modified release dosage form can include, consist of, or consist essentially of 25-hydroxyvitamin D3.


For the compositions and methods described herein, preferred steps, preferred components, preferred compositional ranges thereof, and preferred combinations of the foregoing, can be selected from the various examples provided herein.


Further aspects and advantages will be apparent to those of ordinary skill in the art from a review of the following detailed description, taken in conjunction with the drawings.





BRIEF DESCRIPTION OF THE DRAWINGS

For further facilitating the understanding of the present invention, thirty-one drawing figures are appended hereto.



FIG. 1 through FIG. 8 show plots of the change in serum 25-hydroxyvitamin D3 levels over the first 24 hours post-administration for groups of test subjects administered oral dosage formulations including 25-hydroxyvitamin D3 according to Example 1. In addition, FIG. 7 shows an overlay of comparative data for immediate and controlled release formulations.



FIG. 9 through FIG. 11 show plots of the change in serum 25-hydroxyvitamin D3 levels over the period of the study of Example 1 for the Group 7 controlled release formulation, the Group 9 immediate release formulation according to the prior art, and Group 10 intravenous administration.



FIG. 12 shows an overlay plot of the data in FIG. 9 and FIG. 10 for Groups 7 and 9, respectively, in Example 1.



FIG. 13 through FIG. 18 show mean pharmacokinetic profile for miniature swine dosed with modified and immediate release oral formulations of 25-hydroxyvitamin D3 according to Example 2.



FIG. 19 shows a comparison of pharmacokinetic profiles for MR and IR formulations of 250 μg 25-hydroxyvitamin D3 according to Example 2.



FIG. 20 shows mean uncorrected serum 25-hydroxyvitamin D3 concentration versus time profiles for Groups 1 to 3 of miniature swine after administration of 25-hydroxyvitamin D3 according to Example 3.



FIG. 21 through FIG. 23 show mean baseline corrected serum 25-hydroxyvitamin D3 concentration versus time profiles for Groups 1 to 3 according to Example 3.



FIG. 24 shows the mean change in parathyroid hormone levels for Group 1 animals from predose to day 21, and FIG. 25 shows the mean change in parathyroid hormone levels for Group 2 animals from predose to day 21, from Example 3.



FIG. 26 shows mean serum 25-hydroxyvitamin D3 concentration versus time profiles for Groups 1 to 5 of Beagle dogs administered 25-hydroxyvitamin D3 modified release capsules according to Example 4.



FIG. 27 shows a dissolution release profile for 250 μg capsules according to Example 2, which showed an average release of about 72% of 25-hydroxyvitamin D3 at 24 hours.



FIG. 28 shows a plot of the change in serum 25-hydroxyvitamin D3 levels over 96 hours in Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were intravenously administered a single dose of about 450 μg (specifically, 448 μg) of 25-hydroxyvitamin D3 (N=8) or orally administered a single dose of 450 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8) or orally administered a single dose of 900 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8). The orally administered doses of controlled release 25-hydroxyvitamin D3 gradually increased levels of serum 25-hydroxyvitamin D3, sustained this increase for over 96 hours, and avoided a surge in 25-hydroxyvitamin D3 serum levels.



FIG. 29 shows a plot of the change in serum 1,25-hydroxyvitamin D3 levels over 96 hours in Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were intravenously administered a single dose of about 450 μg (specifically, 448 μg) of 25-hydroxyvitamin D3 (N=8) or orally administered a single dose of 450 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8) or orally administered a single dose of 900 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8). The orally administered doses of controlled release 25-hydroxyvitamin D3 gradually increased levels of serum 1,25-hydroxyvitamin D3, sustained this increase for over 96 hours, and avoided a surge in 1,25-hydroxyvitamin D3 serum levels. The orally administered dose having 900 μg of 25-hydroxyvitamin D3 increased levels of serum 1,25-hydroxyvitamin D3 to a greater extent than the orally administered dose containing 450 μg of 25-hydroxyvitamin D3, which increased 1,25-hydroxyvitamin D3 by only about 2.5 pg/mL.



FIG. 30 shows the mean % change in parathyroid hormone levels for Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were orally administered a single dose of 900 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8), or intravenously administered a single dose of about 450 μg (448 μg) of 25-hydroxyvitamin D3 (N=8). The orally administered single dose of 25-hydroxyvitamin D3 caused an immediate and sustained reduction in mean plasma iPTH, which achieved a maximum reduction of approximately 65% of that achieved following intravenous administration, while the intravenously administered single dose of 25-hydroxyvitamin D3 had minimal to no effect on plasma iPTH. Despite higher levels of 25-hydroxyvitamin D3 and 1,25-hydroxyvitamin D3 in subjects that were intravenously administered 25-hydroxyvitamin D3, greater mean reductions in iPTH occurred in subjects that were orally administered 25-hydroxyvitamin D3 in a modified release dosage form as described herein.



FIG. 31 shows the mean % change in iPTH levels for Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were orally administered a single dose of 900 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8), 450 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8), or intravenously administered a single dose of about 450 μg (specifically 448 μg) of 25-hydroxyvitamin D3 (N=8). The serum calcium levels remained within the normal range throughout the studies demonstrated in FIG. 28 through FIG. 31. In addition, no serious adverse events were reported. The modified release dosage formulation used in the studies underlying the data shown in FIG. 28 through FIG. 31 was the same as formulation 7 in Table 2 herein, with the following exceptions: (1) a vegetable-based capsule was used in place of a gelatin capsule; (2) hard paraffin content was reduced to 20%; and (3) liquid paraffin (light mineral oil) content was increased to 30%. The amount of 25-hydroxyvitamin D3 per capsule was 90 μg. The bioavailability was estimated to be about 7% measured over the first 24 hours post-dose.





DETAILED DESCRIPTION

While the compositions and methods described and claimed herein are susceptible of embodiments in various forms, the description hereafter includes aspects of various embodiments with the understanding that the disclosure is illustrative, and is not intended to limit the invention to the embodiments described herein. It will be appreciated by the skilled artisan that various of the aspects or embodiments described herein are non-exclusive, such that the features of one embodiment can be optionally combined with the features of another embodiment, and all such combinations are contemplated for use in the methods and compositions of the present disclosure.


As used herein, the term “Vitamin D toxicity” is meant to refer to the side effects suffered from excessively elevated Vitamin D blood levels, including one or more of nausea, vomiting, polyuria, hypercalciuria, hypercalcemia and hyperphosphatemia.


“Vitamin D insufficiency and deficiency” is generally defined as having serum 25-hydroxyvitamin D levels below 30 ng/mL (see National Kidney Foundation guidelines, NKF, Am. J. Kidney Dis. 42:S1-S202 (2003), incorporated herein by reference).


As used herein the term “hypercalcemia” refers to a condition in a patient wherein the patient has corrected serum levels of calcium above 10.2 mg/dL. Normal corrected serum levels of calcium for a human are between about 8.6 to 10.2 mg/dL.


As used herein the term “hyperphosphatemia” refers to a condition in a patient having normal kidney function, or Stage 3-4 CKD, wherein the patient has serum phosphorous levels above 4.6 mg/dL. In a patient who has Stage 5 CKD, hyperphosphatemia occurs when the patient has serum levels above 5.5 mg/dL. Normal values for serum phosphorous in a human are 2.5-4.5 mg/dL.


As used herein the term “over suppression of plasma iPTH” refers to a condition in a patient having normal kidney function, or Stage 1-3 CKD, wherein the patient has levels of plasma iPTH below 15 pg/mL. In a patient having Stage 4 CKD, over suppression of plasma iPTH occurs when the patient has levels of plasma iPTH below 30 pg/mL. In a patient having Stage 5 CKD, over suppression of plasma iPTH occurs when the patient has levels of plasma iPTH below 100 pg/mL.


As used herein, the term “Vitamin D hormone replacement therapy” refers to the administration to a patient of an effective amount of an active vitamin D hormone such as 1,25-dihydroxyvitamin D3 and/or 1,25-dihydroxyvitamin D2, optionally together with or other metabolites and analogs of Vitamin D which can substantially occupy the intracellular VDR.


The term “substantially constant” with respect to the serum or blood level of 25-hydroxyvitamin D prohormones means that the release profile of any formulation administered as detailed hereinbelow should not include transient increases in total serum or blood levels of 25-hydroxyvitamin D of greater than approximately 3 ng/mL within 3 hours after administration of a unit dose, optionally not greater than about 4 ng/ml in the first 4 hours, or about 4.4 ng/ml in the first 4 hours, or not greater than about 5 ng/ml in the first 4 hours, or about 6 ng/ml in the first 4 hours after administration of a unit dose. The term “substantially constant” with respect to the serum or blood level of an active vitamin D hormone preferably means that the release profile of the controlled release formulation should not include increases in total serum or blood levels of 1,25-dihydroxyvitamin D of greater than 10 pg/ml each after administration of a unit dose, optionally not greater than 8 pg/mL, optionally over a period of the initial 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, or 10 hours post-dose.


As used herein, the terms “controlled release,” “sustained release,” and “modified release” are used interchangeably. Such dosage forms release the administered vitamin D compound in a way that deviates from immediate release. A sustained release formulation releases 25-hydroxyvitamin D from the formulation over time, such that it is absorbed at various levels in the gastrointestinal tract. For example, the release of a 25-hydroxyvitamin D compound will preferably be at such a rate that total serum or blood levels of 25-hydroxyvitamin D are maintained or elevated above predosing levels for an extended period of time, e.g. 24, 48, or 96 hours, or even longer. The terms optionally also include delayed release characteristics. For example, a delayed, sustained release type of formulation can include a coating such as an enteric coating such that the formulation substantially releases the 25-hydroxyvitamin D in the intestines, or a relatively specific part of the intestines, rather than the stomach.


“Supraphysiologic” in reference to intraluminal, intracellular and blood levels of Vitamin D refers to a total concentration of the vitamin D compound markedly greater than the generally stable levels observed in a Vitamin D-replete subject, animal or human patient over the course of any 24-hour period by laboratory measurement when Vitamin D supplementation has been withheld for at least 30 days. “Adverse supraphysiologic surge” refers to a local or serum concentration of a vitamin D compound that elicits adverse effects such as excessive extrarenal hormone production, leading to local adverse effects on calcium and phosphorus metabolism, inhibition of hepatic 25-hydroxylation of vitamin D, increased catabolism of both Vitamin D and 25-hydroxyvitamin D, hypercalciuria, hypercalcemia and/or hyperphosphatemia, with possible cardiovascular sequelae.


As used herein, the term “hyperparathyroidism” refers to primary hyperparathyroidism, secondary hyperparathyroidism and hyperparathyroidism secondary to chronic kidney disease (Stage 3, Stage 4, or Stage 5).


The term “subject” as used herein generally includes humans, mammals (e.g., dogs, cats, rodents, sheep, horses, cows, goats), veterinary animals and zoo animals.


It also is specifically understood that any numerical value recited herein includes all values from the lower value to the upper value, i.e., all possible combinations of numerical values between the lowest value and the highest value enumerated are to be considered to be expressly stated in this application. For example, if a concentration range or a beneficial effect range is stated as 1% to 50%, it is intended that values such as 2% to 40%, 10% to 30%, or 1% to 3%, etc., are expressly enumerated in this specification. These are only examples of what is specifically intended.


Administration of 25-hydroxyvitamin D3 in an immediate release oral formulation has been tried as an alternative method of Vitamin D supplementation. This approach, which was subsequently abandoned, caused problems as do the currently used Vitamin D supplements. Specifically, it produced surges or spikes in blood and intracellular 25-hydroxyvitamin D levels. Without intending to be bound by any particular theory, it is believed that surges or spikes in blood and intracellular 25-hydroxyvitamin D levels promote (a) competitive displacement of Vitamin D hormones from the serum Vitamin D Binding Protein (DBP) and excessive delivery of the displaced hormones to tissues containing VDR, and (b) transiently excessive renal and extrarenal production of Vitamin D hormones, which together led to local aberrations in calcium and phosphorus metabolism. In addition, these surges in blood 25-hydroxyvitamin D levels are believed to promote catabolism of both Vitamin D and 25-hydroxyvitamin D by 24- and/or 26-hydroxylation in the kidney and other tissues, down-regulation of hepatic production of Vitamin D prohormones, unnecessarily impeding the efficient repletion of Vitamin D insufficiency or deficiency, and, additional local aberrations in calcium and phosphorus homeostasis mediated by direct binding to VDR. Importantly, immediate release of 25-hydroxyvitamin D3 is believed to promote its intestinal absorption via a mechanism substantially involving transport to the liver in chylomicrons, rather than bound to the serum DBP. Delivery of 25-hydroxyvitamin D to the liver via chylomicrons is believed to significantly increase the likelihood of its catabolism.


One aspect of the disclosure provides a solid or semi-solid, waxy pharmaceutical formulation for controlled release of 25-hydroxyvitamin D in the gastrointestinal tract of a subject which ingests the formulation. The formulation includes a waxy controlled release carrier agent, a lipoidic agent, an oily vehicle for 25-hydroxyitamin D, and 25-hydroxyvitamin D. The formulation provides for controlled release of 25-hydroxyvitamin D incorporated therein. The formulation can be free of or essentially free of disintegrants.


The waxy controlled release carrier provides for a formulation which is solid or in the alternative semi-solid at room temperature. The formulation can be solid, semi-solid, or liquid at body temperature. In one embodiment, the formulation will be semi-solid at body temperature. In another embodiment, the formulation will be liquid at body temperature. In another embodiment, the formulation will be solid at body temperature. Examples of carriers suitable for use include waxes, such as synthetic wax, microcrystalline wax, paraffin wax, carnauba wax, and beeswax; polyethoxylated castor oil derivatives, hydrogenated vegetable oils, glyceryl mono-, di- or tribehenates; long-chain alcohols, such as stearyl alcohol, cetyl alcohol, and polyethylene glycol; and mixtures of any of the foregoing. Non-digestible waxy substances, such as hard paraffin wax, are preferred.


The waxy carrier preferably is present in an amount greater than about 5% of the formulation, based on the total weight of the formulation excluding any additional coatings or shells (wt %). For example, the waxy carrier can comprise greater than 5 wt % of the formulation, greater than 10 wt % of the formulation, greater than 15 wt % of the formulation, greater than 20 wt % of the formulation, and greater than 25 wt % of the formulation. The waxy carrier is preferably present in an amount less than 50 wt %, less than 40 wt %, less than 35 wt %, or less than 30 wt. %. Suitable ranges include 5 wt % to 35 wt %, 15 wt % to 35 wt % and 20 to 30 wt %. Examples include 15 wt %, 16 wt %, 17 wt %, 18 wt %, 19 wt %, 20 wt %, 21 wt %, 22 wt %, 23 wt %, 24 wt %, 25 wt %, 26 wt %, 27 wt %, 28 wt %, 29 wt %, and 30 wt %.


The lipoidic agent provides for release of 25-hydroxyvitamin D from the formulation in the gastrointestinal tract of the subject being treated. Without intending to be bound by any particular theory of operation, it is believed that the lipoidic agent can serve one or more preferred functions such as creating a micro-emulsion of the oily vehicle in gastrointestinal fluid; providing prolonged gastric retention, for example by bioadhesive properties such that the formulation interacts with the mucous layer of the stomach and/or intestine; and in enhancing absorption of 25-hydroxyvitamin D. However, regardless of the mechanism of action, the invention is not limited by any particular mode of operation.


The lipoidic agent components preferably are amphiphiles, in which the molecule or ion contains both hydrophilic and lipophilic portions. These components can be defined by a numerical value based on the Hydrophile/Lipophile Balance system (“HLB system”). The HLB scale is a numerical scale, extending from 0 to approximately 20, where lower numbers denote more lipophilic and hydrophobic substances, and higher numbers denote more hydrophilic and lipophobic substances. The affinity of a compound for water, or for oily substances, is determined and its HLB value is assigned experimentally. The HLB of the hydrophobic carrier employed herein preferably will be in a range of about 13 to about 18.


A variety of pharmaceutically acceptable lipoidic agents may be incorporated in the formulation. The quantity of lipoidic agent present in the formulation is preferably at least 5 wt %, at least 15 wt %, at least 35 wt %, at least 40 wt % or at least 45 wt %. Suitable ranges include about 5 wt % to about 60 wt %, about 20 wt % to about 60 wt % and about 40 wt % to about 50 wt %.


In one embodiment, the lipoidic agent is a lipophilic emulsifier which has an HLB of less than 7 and comprises a member selected from the group consisting of mixed fatty acid monoglycerides; mixed fatty acid diglycerides; mixtures of fatty acid mono- and diglycerides; lipophilic polyglycerol esters; glycerol esters including glyceryl monooleate, glyceryl dioleate, glyceryl monostearate, glyceryl distearate, glyceryl monopalmitate, and glyceryl dipalmitate; glyceryl-lacto esters of fatty acids; propylene glycol esters including propylene glycol monopalmitate, propylene glycol monostearate, and propylene glycol monooleate; sorbitan esters including sorbitan monostearate, sorbitan sesquioleate; fatty acids and their soaps including stearic acid, palmitic acid, and oleic acid; and mixtures thereof glyceryl monooleate, glyceryl dioleate, glyceryl monostearate, glyceryl distearate, glyceryl monopalmitate, and glyceryl dipalmitate; glyceryl-lacto esters of fatty acids; propylene glycol esters including propylene glycol monopalmitate, propylene glycol monostearate, and propylene glycol monooleate; sorbitan esters including sorbitan monostearate, sorbitan sesquioleate; fatty acids and their soaps including stearic acid, palmitic acid, and oleic acid; and mixtures thereof.


A preferred lipoidic agent is selected from glycerides and derivatives thereof. Preferred glycerides are selected from the group consisting of medium or long chain glycerides, caprylocaproyl macrogolglycerides, and mixtures thereof.


Preferred medium chain glycerides include, but are not limited to, medium chain monoglycerides, medium chain diglycerides, caprylic/capric triglyceride, glyceryl monolaurate, glyceryl monostearate, caprylic/capric glycerides, glycerylmonocaprylate, glyceryl monodicaprylate, caprylic/capric linoleic triglyceride, and caprylic/capric/succinic triglyceride.


Monoglycerides having a low melting point are preferred for making the formulation. Preferred monoglycerides include but are not limited to, glyceryl monostearate, glyceryl monopalmitate, glyceryl monooleate, glyceryl monocaprylate, glyceryl monocaprate, glyceryl monolaurate, etc., preferably glyceryl monostearate (GMS). GMS is a natural emulsifying agent. It is oil soluble, but poorly soluble in water. GMS has an HLB value of 3.8. Another preferred monoglyceride is glyceryl monooleate (GMO). GMO is also a natural emulsifying agent; it is oil soluble, but poorly soluble in water, and it has an HLB value of 3.8.


In another embodiment, the glyceride is an absorption enhancer selected from caprylocaproyl macrogolglycerides. Caprylocaproyl macrogolglycerides which may be employed include, but are not limited to, polyethylene glycosylated glycerides, also known as polyglycolized glycerides or PEGylated glycerides. PEGylaed glycerides which may be employed in the composition include, but are not limited to, mixtures of monoglycerides, diglycerides, and triglycerides and monoesters and diesters of polyethylene glycol, polyethylene glycosylated almond glycerides, polyethylene glycosylated corn glycerides, and polyethylene glycosylated caprylic/capric triglyceride. The absorption enhancer preferably has an HLB value from 13 to 18, more preferably from 13 to 15.


One preferred absorption enhancer is known under the trade name GELUCIRE, and is commercially available from Gattefossé Corporation, Paramus, N.J., USA. GELUCIRE is a well known excipient which is a family of fatty acid esters of glycerol and PEG esters, also known as polyglycolized glycerides. GELUCIRE is used in various applications including preparing sustained release pharmaceutical compositions. GELUCIRE compounds are inert, semi-solid waxy materials which are amphiphilic and are available with varying physical characteristics such as melting point, HLB, and solubilities in various solvents. They are surface active in nature and disperse or solubilize in aqueous media forming micelles, microscopic globules or vesicles. They are identified by their melting point/HLB value. The melting point is expressed in degrees Celsius. One or a mixture of different grades of GELUCIRE excipient may be chosen to achieve the desired characteristics of melting point and/or HLB value. The preferred GELUCIRE composition is GELUCIRE 44/14, a semisolid waxy material with a melting point of 44° C. and a HLB of 14.


Another preferred polyglycolyzed glyceride absorption enhancer is caprylocaproyl macrogol-8-glyceride (CAS No. 85536-07-8 and 84963-88-2). This is a mixture of mono-, di- and triesters of glycerol and of PEG 400 with medium-chain fatty acids (C8-C10) which is marketed, for example, by Gattefossé Corporation, Paramus, N.J., USA under the trade name LABRASOL. LABRASOL has a HLB value of 14 and has the following composition by weight: C8-C10 monoglycerides approximately 4%; C8-C10 diglycerides approximately 17%; C8-C10 triglycerides approximately 6%; C8-C10 monoesters of PEG 400 approximately 14%; C8-C10 diesters of PEG 400 approximately 36%; free PEG 400 approximately 20%; free glycerol approximately 3%.


Preferably, the lipoidic agent includes a mixture of a lipophilic emulsifier which has an HLB of less than 7 and an absorption enhancer that preferably has an HLB value from 13 to 18. The lipophilic emulsifier is preferably present in an amount in a range of about 20 wt % to about 50 wt %, preferably about 30 wt % to about 40 wt %, and the absorption enhancer is preferably present in an amount of about 5 to about 20 wt %, preferably about 8 to about 15 wt %.


The low melting points of many of the solid lipoidic compositions provide a means of incorporating the pharmaceutically active ingredients in them at temperatures from about 0° C. to about 50° C. above their respective melting points, and then filling the melt (solution and/or dispersion) in animal or vegetable gelatin capsules. The melt solidifies inside the capsules upon cooling to room temperature.


The oily component serves as a vehicle, preferably the main vehicle, for 25-hydroxyvitamin D. Any pharmaceutically-acceptable oil can be used. Examples include animal (e.g., fish), vegetable (e.g., soybean), and mineral oils. The oil preferably will readily dissolve 25-hydroxyvitamin D. Preferred oily components include non-digestible oils, such as mineral oils, particularly liquid paraffins, and squalene. The oil vehicle preferably comprises about 10 wt % to about 50 wt % of the formulation, more preferably about 15 wt % to about 45 wt % about 20 wt % to about 40 wt %, or about 15 wt % to about 25 wt %. In one preferred embodiment, the liquid paraffin can be characterized by one or more of the following parameters: specific gravity about 0.88 to 0.89; kinematic viscosity (40° C.) about 64 to about 70 cSt; molecular weight 424; % paraffinic hydrocarbons about 59; and pour point −24° C. The ratio between the waxy component and the oily component can be optimized in order to achieve the desired rate of release of 25-hydroxyvitamin D. Thus, if a heavier oil component is used, relatively less of the waxy component can be used, and if a lighter oil component is used, then relatively more waxy component can be used.


25-hydroxyvitamin D compounds suitable for prophylactic and/or therapeutic use as described herein include 25-hydroxyvitamin D3, 25-hydroxyvitamin D2, analogs thereof, and combinations of any of the foregoing. These compounds and combinations are generally referred to herein under the generic term 25-hydroxyvitamin D. In one type of embodiment, 25-hydroxyvitamin D includes one or more hydroxy forms, such as a combination of 25-hydroxyvitamin D3 and 25-hydroxyvitamin D2. In embodiments, 25-hydroxyvitamin D can be administered in a therapeutically effective amount (e.g., amount effective to prevent or treat a condition described herein, for example, hypovitaminosis D and/or hyperparathyroidism).


The invention also includes modified release compositions comprising oral formulations of 25-hydroxyvitamin D and methods of administering such formulations to treat hyperparathyroidism. Optionally, the methods will have one or more advantages including avoiding supraphysiological surges in intraluminal, intracellular and blood levels of 25-hydroxyvitamin D and their consequences; avoiding substantially increased catabolism of the administered 25-hydroxyvitamin D; and, avoiding serious side effects associated with Vitamin D supplementation, namely Vitamin D toxicity.


The controlled release compositions intended for oral administration in accordance with the methods describe herein preferably are designed to contain concentrations of the 25-hydroxyvitamin D of 1 to 1000 μg per unit dose and are prepared and delivered in such a manner as to effect controlled or substantially constant release of the 25-hydroxyvitamin D, optionally into the ileum of the gastrointestinal tract, of humans or animals over an extended period of time. Preferred dosages include 1 to 1000 μg per unit dose, 1 to 600 μg, 1 to 400 μg, 1 to 200 μg, 1 to 100 μg, 5 to 90 μg, 30 to 80 μg, 20 to 60 μg, 30 to 60 μg, 35 to 50 μg, 5 to 50 μg, and 10 to 25 μg, for example 20 μg, 25 μg, 30 μg, 40 μg, 50 μg, 60 μg, 70 μg, 80 μg, 90 μg, and 100 μg. The compositions and methods may provide substantially increased absorption of 25-hydroxyvitamin D via transport on DBP and decreased absorption via transport in chylomicrons. The compositions and methods may provide maintenance of substantially constant blood levels of 25-hydroxyvitamin D during the 24-hour post-dosing period, and preferably 168 hours post-dosing, and further preferably to 720 hours post-dosing. By providing a gradual, sustained and direct release of the 25-hydroxyvitamin D and absorption preferentially to circulating DBP (rather than to chylomicrons), blood, intraluminal and intracellular 25-hydroxyvitamin D concentration spikes, i.e., supraphysiologic levels, and related unwanted catabolism can be mitigated or eliminated. Furthermore, by providing a gradual and sustained release, serum levels of 25-hydroxyvitamin D can be increased and maintained more predictably and reliably than by administration of immediate release oral formulations, allowing for a consistent dosage and reducing or eliminating the need for frequent patient monitoring.


It will be recognized that such dosage forms can have bioavailabilities less than 100%. Accordingly, viewed one way a dosage amount is a nominal dosage, not accounting for bioavailability, and viewed another way a dosage amount is the effective dosage amount, accounting for bioavailability.


In one preferred class of embodiments, the modified release formulation releases at least 5-80%, for example at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, or at least 72% of 25-hydroxyvitamin D within the first 24 hours after dosing.


The invention also contemplates that doses of 25-hydroxyvitamin D may be given less frequently than once per day, for example at intervals of once every other day, three times a week, twice a week, weekly, every 2 weeks, every 3 weeks, every 4 weeks, every 5 weeks, or every 6 weeks, for example, wherein higher effective doses generally permit for less frequent dosing and lower effective doses generally permit for more frequent dosing.


Advantageously, 25-hydroxyvitamin D together with other optional therapeutic agents can be orally administered in accordance with the above described embodiments in nominal dosage amounts equivalent to a range of 1 to 180 μg per day, with the preferred dosage amounts in a range of 30 to 120 μg per day, for example about 60 to 90 μg. Examples contemplated include 30 μg per day, 60 μg per day, 90 μg per day, 120 μg per day, and 150 μg per day. Viewed from an effective dose perspective, accounting for bioavailability, it is contemplated that one type of preferred dosage amount will be in a range of about 2 μg per day to about 8 μg per day, or about 4 μg per day to about 7 μg per day, for example about 2 μg, about 4 μg, about 6 μg, 8.5 μg, or 10.5 μg effective dose per day based on bioavailability of 7% in the first 24 hours following dosing. For a formulation having 10% bioavailability calculated over a range of 42 days post-dose, preferred effective doses would be in a range of 3 to 12 μg per day, for example about 6 to 9 μg. Examples contemplated include 3 μg per day, 6 μg per day, 9 μg per day, 12 μg per day, and 15 μg per day.


In one aspect, the 25-hydroxyvitamin D can be orally administered in accordance with the above described embodiments in nominal dosage amounts of at least 30 μg, or at least 300 μg, or at least 350 μg, or at least 400 μg, for example in a range of 30 μg to 1800 μg, or 450 μg to 1800 μg, or 30 μg to 1000 μg and on a frequency less than once daily, for example every other day, every third day, every fourth day, weekly, every two weeks, every three weeks, every four weeks, monthly, every five weeks, or every six weeks. Viewed from an effective dose perspective, accounting for bioavailability, a range of about 20 μg to 400 μg is contemplated, and can be delivered on a frequency less than once daily, for example every other day, every third day, every fourth day, weekly, every two weeks, every three weeks, every four weeks, monthly, every five weeks, or every six weeks. From an effective dose perspective, accounting for bioavailability, it is contemplated that one type of preferred dosage amount for less frequent dosing will be in a range of about 30 μg to about 130 μg, based on bioavailability in the first 24 hours following dosing. For a formulation having 10% bioavailability calculated over a range of 42 days post-dose, preferred effective doses would be greater than 45 μg, at least 50 μg, at least 60 μg, at least 70 μg, at least 80 μg, or at least 90 μg, for example in a range of 50 μg to 180 μg or a range of 70 μg to 110 μg. Preferred dosing regimens (amount and frequency) will provide an average rise in serum 25-hydroxyvitamin D3 of about 1 to 20 ng/mL at 30 days post-dose. It will be understood that the aforementioned dosage amounts described in equivalent amounts per day will be administered according to the methods described herein in intervals longer than once per day; thus, for example, a nominal dose equivalent of 100 μg per day can be administered as 200 μg every other day, or 700 μg per week.


In any of the foregoing methods the daily dose described can be delivered on a less frequent dosing regimen to result in an average daily dose within the ranges described above.


In one type of embodiment, a preferred dose will provide an maximum rise in serum total 25-hydroxyvitamin D of greater than 6 ng/mL, or at least 7 ng/mL, or at least 8 ng/mL, or at least 9 ng/mL, or at least 10 ng/mL, or at least 11 ng/mL, or at least 12 ng/mL, or at least 14 ng/mL, or at least 16 ng/mL, and at most 30 ng/mL, or 25 ng/mL, or 20 ng/mL, or 15 ng/mL, for example in a range of 7 to 30 ng/mL, or 8 to 16 ng/mL, or 10 to 14 ng/mL, for example 12 ng/mL within 24 hours post-dose in a dose interval. Optionally, the maximum rise just described can be measured within 12 hours post-dose. Still further optionally, the limit on rise in serum total 25-hydroxyvitamin D achieved by the method will conform to the 48-hours post dose period, or also the 96-hours post-dose period.


In another type of embodiment, a preferred dose will provide an average rise in serum 25-hydroxyvitamin D of at least about 7 ng/mL, or in a range of about 7 to 14 ng/mL within 48 hours post-dose in a dose interval. In another type of embodiment, a preferred dose will provide an average rise in serum total 25-hydroxyvitamin D in a range of about 7 to 30 ng/mL within 48 hours post-dose in a dose interval.


In embodiments, the method is contemplated to include administering a formulation described herein to raise and preferably also maintain blood 1,25-dihydroxyvitamin D levels at 25 pg/mL, 30 pg/mL, or higher, e.g. 25-65 pg/mL for an extended period, for example at least one month, at least three months, at least six months, or longer. In another type of embodiment, a preferred dose will provide an average rise in serum total 1,25-dihydroxyvitamin D of greater than about 3 pg/mL and less than about 10 pg/mL within the first 24 hours of dosing, or at least about 7 pg/mL and less than about 10 pg/mL within the first 48 hours post-dose in a dose interval.


In one aspect, a method for lowering or maintaining lowered serum parathyroid hormone in human patients includes administering to said patients an effective amount of a modified release dosage form comprising 25-hydroxyvitamin D to lower or maintain lowered serum parathyroid hormone levels, preferably an amount that lowers iPTH levels by at least 10%, 15%, 20%, 25%, or 30%, compared to baseline, or alternatively the amount needed to reduce serum levels of PTH to the target range for the CKD stage (e.g., for Stage 3 is 35-70 pg/mL (equivalent to 3.85-7.7 pmol/L), for Stage 4 is 70-110 pg/mL (equivalent to 7.7-12.1 pmol/L), and for Stage 5 is 150-300 pg/mL (equivalent to 16.5-33.0 pmol/L) (defined in K/DOQI Guideline No. 1)). In embodiments, the patient's iPTH remains lowered for at least 48 hours, at least 60 hours, at least 72 hours, at least 4 days, at least 5 days, at least 6 days, or at least 7 days post-dose in a dose interval.


In another aspect, a method for lowering or maintaining lowered serum parathyroid hormone in human patients includes administering to said patients an effective amount of a modified release dosage form comprising 25-hydroxyvitamin D according to the disclosure herein to lower or maintain lowered serum parathyroid hormone levels, to within K-DIGO guidelines (i.e. for Stages 3, 4, or 5 CKD patients not on dialysis, one times the upper limit of normal for the assay used; for Stage 5 CKD patients on dialysis, about two to nine times the assay's upper-normal limit, or about 130 pg/mL to 600 pg/mL), or in the alternative to 65 pg/mL iPTH or less.


As described above, one aspect of the invention is a method of administering an amount of one or more 25-hydroxyvitamin D compounds to a patient such that the time for the plasma concentration of total 25-hydroxyvitamin D to reach its maximum in a dose interval following administration (Tmax) is increased as compared to Tmax for an equivalent amount of 25-hydroxyvitamin D administered by bolus IV injection. In another embodiment the method includes administering an amount of one or more 25-hydroxyvitamin D compounds to a patient such that the time for the plasma concentration of total 25-hydroxyvitamin D to reach its maximum in a dose interval following administration (Tmax) is increased as compared to Tmax for an equivalent immediate-release, oral dosage form. Practice of a method described herein with a modified release dosage form comprising 25-hydroxyvitamin D preferably will result in a Tmax of serum 25-hydroxyvitamin D at least 4 hours post-dose, or at least 6 hours post-dose, or at least 8 hours post-dose, or at least 10 hours post-dose, or at least 12 hours post-dose, or at least 134 hours post-dose, or greater than 4 hours and up to 48 hours, for example in a range of 6 hours to 24 hours, 8 hours to 20 hours, 10 hours to 18 hours, or 12 hours to 16 hours, for example at least about 13 or 14 hours post-dose.


Similarly, another aspect of the invention is a method of administering an amount of one or more 25-hydroxyvitamin D compounds to a patient such that the time for the plasma concentration of serum total 1,25-dihydroxyvitamin D to reach its maximum in a dose interval following administration (Tmax) is increased as compared to Tmax for an equivalent amount of 25-hydroxyvitamin D administered by bolus IV injection. In an alternative embodiment the method will involve administering an amount of one or more 25-hydroxyvitamin D compounds to a patient such that the time for the plasma concentration of serum total 1,25-dihydroxyvitamin D to reach its maximum in a dose interval following administration (Tmax) is increased as compared to Tmax for an equivalent immediate-release, oral dosage form. Practice of a method described herein with a modified release dosage form comprising 25-hydroxyvitamin D preferably will result in a Tmax of serum 1,25-dihydroxyvitamin D at least 100 hours post-dose, or at least 110 hours post-dose, or at least 120 hours post-dose, or at least 130 hours post-dose, or at least 140 hours post-dose, or at least 150 hours post-dose, or at least 160 hours post-dose, or at least 170 hours post-dose, or at least 180 hours post-dose, or at least 190 hours post-dose, or greater than 100 hours and up to 300 hours, for example in a range of 120 hours to 240 hours, 140 hours to 220 hours, or 160 hours to 200 hours, for example about 180 or 190 hours post-dose.


The dosages described herein are contemplated for any of the therapeutic methods described herein. It will be appreciated that the actual preferred amount of 25-hydroxyvitamin D in a specific case will vary according the particular compositions formulated, the mode of application, and the particular situs being treated. Dosages can be determined using conventional considerations, e.g., by customary comparison of the differential activity of the hormone and of a known agent, e.g. by means of an appropriate conventional pharmacological protocol.


The specific doses for each particular patient can depend on a wide variety of factors, for example, on the age, body weight, general state of health, sex, on the diet, on the timing and mode of administration, on the rate of excretion, and on medicaments used in combination and the severity of the particular disorder to which the therapy is applied.


Patients in need of vitamin D supplementation include healthy subjects and subjects at risk for vitamin D insufficiency or deficiency, for example, subjects with Stage 1, 2, 3, 4 or 5 CKD; infants, children and adults that do not drink vitamin D fortified milk (e.g. lactose intolerant subjects, subjects with milk allergy, vegetarians who do not consume milk, and breast fed infants); subjects with rickets; subjects with dark skin (e.g., in the U.S., 42% of African American women between 15 and 49 years of age were vitamin D deficient compared to 4% of white women); the elderly (who have a reduced ability to synthesize vitamin D and also are more likely to stay indoors); institutionalized adults (who are likely to stay indoors, including subjects with Alzheimer's disease or mentally ill); subjects who cover all exposed skin (such as members of certain religions or cultures); subjects who always use sunscreen (e.g., the application of sunscreen with a Sun Protection Factor (SPF) value of 8 reduces production of vitamin D by 95%, and higher SPF values may further reduce vitamin D); subjects with fat malabsorption syndromes (including but not limited to cystic fibrosis, cholestatic liver disease, other liver disease, gallbladder disease, pancreatic enzyme deficiency, Crohn's disease, inflammatory bowel disease, sprue or celiac disease, or surgical removal of part or all of the stomach and/or intestines); subjects with inflammatory bowel disease; subjects with Crohn's disease; subjects who have had small bowel resections; subjects with gum disease; subjects taking medications that increase the catabolism of vitamin D, including phenyloin, fosphenyloin, phenobarbital, carbamazepine, and rifampin; subjects taking medications that reduce absorption of vitamin D, including cholestyramine, colestipol, orlistat, mineral oil, and fat substitutes; subjects taking medications that inhibit activation of vitamin D, including ketoconazole; subjects taking medications that decrease calcium absorption, including corticosteroids; subjects with obesity (vitamin D deposited in body fat stores is less bioavailable); subjects with osteoporosis; and/or postmenopausal women. According to the Institute of Medicine's report on the Dietary Reference Intakes for vitamin D, food consumption data suggest that median intakes of vitamin D for both younger and older women are below current recommendations; data suggest that more than 50% of younger and older women are not consuming recommended amounts of vitamin D.


Optionally excluded from the methods of the invention described herein are therapeutic treatment of subjects suffering from renal osteodystrophy (including osteomalacia and osteitis fibrosa cystica).


In other aspects, the compositions and methods of the invention are useful for prophylactic or therapeutic treatment of vitamin D-responsive diseases, i.e., diseases where vitamin D, 25-hydroxyvitamin D or active vitamin D (e.g., 1,25-dihydroxyvitamin D) prevents onset or progression of disease, or reduces signs or symptoms of disease. Such vitamin D-responsive diseases include cancer (e.g., breast, lung, skin, melanoma, colon, colorectal, rectal, prostate and bone cancer). 1,25-dihydroxyvitamin D has been observed to induce cell differentiation and/or inhibit cell proliferation in vitro for a number of cells. Vitamin D-responsive diseases also include autoimmune diseases, for example, type I diabetes, multiple sclerosis, rheumatoid arthritis, polymyositis, dermatomyositis, scleroderma, fibrosis, Grave's disease, Hashimoto's disease, acute or chronic transplant rejection, acute or chronic graft versus host disease, inflammatory bowel disease, Crohn's disease, systemic lupus erythematosis, Sjogren's Syndrome, eczema and psoriasis, dermatitis, including atopic dermatitis, contact dermatitis, allergic dermatitis and/or chronic dermatitis. Vitamin D-responsive diseases also include other inflammatory diseases, for example, asthma, chronic obstructive pulmonary disease, polycystic kidney disease, polycystic ovary syndrome, pancreatitis, nephritis, hepatitis, and/or infection. Vitamin D-responsive diseases have also been reported to include hypertension and cardiovascular diseases. Thus, the invention contemplates prophylactic or therapeutic treatment of subjects at risk of or suffering from cardiovascular diseases, for example, subjects with atherosclerosis, arteriosclerosis, coronary artery disease, cerebrovascular disease, peripheral vascular disease, myocardial infarction, myocardial ischemia, cerebral ischemia, stroke, congestive heart failure, cardiomyopathy, obesity or other weight disorders, lipid disorders (e.g. hyperlipidemia, dyslipidemia including associated diabetic dyslipidemia and mixed dyslipidemia hypoalphalipoproteinemia, hypertriglyceridemia, hypercholesterolemia, and low HDL (high density lipoprotein)), metabolic disorders (e.g. Metabolic Syndrome, Type II diabetes mellitus, Type I diabetes mellitus, hyperinsulinemia, impaired glucose tolerance, insulin resistance, diabetic complication including neuropathy, nephropathy, retinopathy, diabetic foot ulcer and cataracts), and/or thrombosis.


Diseases which can benefit from a modulation in the levels of vitamin D compounds, include, but are not limited to: (i) in the parathyroid—hypoparathyroidism, Pseudohypo-parathyroidism, secondary hyperparathyroidism; (ii) in the pancreas—diabetes; (iii) in the thyroid—medullary carcinoma; (iv) in the skin—psoriasis; wound healing; (v) in the lung—sarcoidosis and tuberculosis; (vi) in the kidney—chronic kidney disease, hypophosphatemic VDRR, vitamin D dependent rickets; (vii) in the bone—anticonvulsant treatment, fibrogenisis imperfecta ossium, osteitis fibrosa cystica, osteomalacia, osteoporosis, osteopenia, osteosclerosis, renal osteodytrophy, rickets; (viii) in the intestine—glucocorticoid antagonism, idopathic hypercalcemia, malabsorption syndrome, steatorrhea, tropical sprue; and (ix) autoimmune disorders.


In embodiments of the invention, the disease that benefits from a modulation in the levels of vitamin D compounds are selected from cancer, dermatological disorders (for example psoriasis), parathyroid disorders (for example hyperparathyroidism and secondary hyperparathyroidism), bone disorders (for example osteoporosis) and autoimmune disorders.


A modified release formulation can be prepared by procedures well known to one of ordinary skill in the art. Pharmaceutically acceptable waxes, lipoidic agents, and oils can be melted, if necessary, to provide a flowable liquid thereby making it easier to obtain a homogeneous mixture. 25-Hydroxyvitamin D can be added to the thus liquid carrier, for example dissolved in an alcohol such as anhydrous ethanol, and the ingredients can be mixed to provide a homogeneous mixture. The mixture can be cooled and stored prior to later division into unit dosage forms, such as filled gelatin capsules.


In one preferred method, a portion of the oil vehicle, solid wax, and a lipophilic emulsifier are heated to a relatively high temperature (e.g., 65° C.) and mixed prior to adding an absorption enhancer, followed by additional mixing until homogenous, then cooling to an intermediate elevated temperature (e.g., 50° C. to 55° C.). In a separate vessel, an antioxidant preservative and the remainder of the oil vehicle are mixed and heated to an intermediate elevated temperature (e.g., 50° C.), then combined and mixed with the wax mixture until a homogenous solution is obtained. Next, a solution of 25-hydroxyvitamin D in alcohol is combined with the homogenous waxy solution, mixed until a homogenous solution is obtained, preferably filled into capsules, and then cooled to room temperature. In another preferred method, a portion of the oil vehicle, solid wax, and a lipophilic emulsifier are heated at a temperature of 55° C. to 60° C. and mixed prior to adding an absorption enhancer, followed by additional mixing until homogenous. In a separate vessel, an antioxidant preservative and the remainder of the oil vehicle are mixed and heated to a temperature of 55° C. to 60° C., then combined and mixed with the wax mixture until a homogenous solution is obtained. Next, a solution of 25-hydroxyvitamin D in alcohol is combined with the homogenous waxy solution, mixed until a homogenous solution is obtained, preferably filled into capsules, and then cooled to room temperature.


The formulation preferably is placed in capsules prior to administration to the patient in need of treatment. Such capsules may be hard or soft, and soft capsules are preferred. The formulation may be filled into gelatin capsules using standard capsule filling machinery, such as by melting the formulation and injection filling it into soft capsule shells.


The formulation and methods of use and making are contemplated to include embodiments including any combination of one or more of the additional optional elements, features, and steps further described below, unless stated otherwise.


Thus, in one type of embodiment, the formulation further includes a preservative, such as an antioxidant. Butylated hydroxytoluene (BHT) is preferred.


In another type of embodiment, 25-hydroxyvitamin D is administered in combination with one or more other therapeutic agents.


If 25-hydroxyvitamin D is administered in combination with one or more other therapeutic agents, the proportions of each of the compounds in the combination being administered will be dependent on the particular disease state being addressed. For example, one may choose to orally administer 25-hydroxyvitamin D with one or more calcium salts (intended as a calcium supplement or dietary phosphate binder), bisphosphonates, calcimimetics, nicotinic acid, iron, phosphate binders, cholecalciferol, ergocalciferol, active Vitamin D sterols, glycemic and hypertension control agents, various antineoplastic agents and inhibitors of CYP24 and other cytochrome P450 enzymes that can degrade vitamin D agents. In addition, one may choose to intravenously administer 25-hydroxyvitamin D2 and/or 25-hydroxyvitamin D3 with cholecalciferol, ergocalciferol, active Vitamin D sterols, glycemic and hypertension control agents, various antineoplastic agents and inhibitors of CYP24 and other cytochrome P450 enzymes that can degrade vitamin D agents. In practice, higher doses of the compounds of the present invention are used where therapeutic treatment of a disease state is the desired end, while the lower doses can be used for prophylactic purposes, it being understood that the specific dosage administered in any given case will be adjusted in accordance with the specific compounds being administered, the disease to be treated, the condition of the subject and the other relevant medical facts that may modify the activity of the drug or the response of the subject, as is well known by those skilled in the art.


As described above, the formulation is preferably filled into gelatin capsules, but it may also be administered in neat form, or with one or more external coating layers, such as an enteric coating. It is also contemplated that the formulation can be pressed into tablets, and in such cases one or more tablet pressing excipients may be included.


In the compositions and methods described herein, preferred steps, preferred components, preferred compositional ranges thereof, and preferred combinations of the foregoing, can be selected from the various specific examples provided herein. For example, a preferred formulation includes 25-hydroxyvitamin D (e.g. 25-hydroxyvitamin D3, for example about 0.1 wt % (e.g. 0.12 wt %)), about 2 wt % (e.g., 2.32 wt %) ethanol, about 10 wt % (e.g., 9.75 wt %) GELUCIRE 44/14, about 27 wt % (e.g., 27.51 wt. %) hard paraffin, about 38 wt % (e.g., 37.85 wt %) GMS, about 22 wt % (e.g., 22.43 wt %) mineral oil, and optionally a small amount of preservative (e.g., 0.02 wt % BHT). A variation on this formulation will include about 20% hard paraffin and about 29% mineral oil.


Specifications for still another preferred embodiment of a capsule, and a 50 μg embodiment, are shown in Table 1 below.













TABLE 1








Milligram per




Ingredient
capsule
% w/w




















25-hydroxyvitamin D3
0.040
0.024



Dehydrated ethanol
4.22
2.48



Hard Paraffin
33.97
19.98



Mineral Oil
50.80
29.88



GELUCIRE 44/14
16.59
9.76



GMS
64.35
37.85



BHT
0.034
0.020



Total
170.00
100.00










EXAMPLES

The following Examples illustrate specific formulations and methods for their preparation. The Examples are provided for illustration and are not intended to limit the scope of the invention.


Example 1—Modified Release Formulations

Nine oral vitamin D formulations were prepared according to Table 2 below by homogeneously mixing the identified components in the amounts shown and filling the mixtures into hard gelatin capsules. Formulation 9 is an immediate-release formulation according to the prior art, wherein MIGLYOL 812N is the trade name for caprylic/capric triglycerides, available from CONDEA Chemie GmbH of Cranford, N.J., USA. The formulations were administered to groups of Yucatan miniature swine (about 10 kg), in single doses equivalent to 250 μg of 25-hydroxyvitamin D3. Each group included five animals. An equivalent 250 μg of 25-hydroxyvitamin D3 was administered to a tenth group of five Yucatan miniature swine via intravenous injection.


Blood was collected pre-dose, and at 0.5, 1, 2, 4, 6, 8, 12, 24, 48, 96, 168, 240, 336, 432, 504, 576, and 672 hours post dosing. Serum 25-hydroxyvitamin D3 levels were assayed by Liquid Chromatography/Mass Spectrometry/Mass Spectrometry (LC MS/MS).


Plots of the change in serum 25-hydroxyvitamin D3 levels over the first 24 hours for Groups 1-8 are shown in FIG. 1 through FIG. 8. In addition, the data for the Group 9 immediate release control are plotted with the Group 7 data in FIG. 7. The concentration profiles show that the Group 7 formulation according to the invention (a) produced a gradually increasing and sustained rise in serum 25-hydroxyvitamin D3 levels over the first 24 hours, and (b) avoided a surge in 25-hydroxyvitamin D3 levels.



FIG. 9 through FIG. 11 show plots of the change in serum 25-hydroxyvitamin D3 levels over the period of the study for Groups 7, 9, and 10, respectively. FIG. 12 shows an overlay plot of the data in FIG. 9 and FIG. 10 for Groups 7 and 9, respectively.


The concentration profiles shows that the Group 7 formulation according to the invention produced a gradually increasing rise in serum 25-hydroxyvitamin D3 levels, avoided a surge in 25-hydroxyvitamin D3 levels, and produced a sustained increase of serum 25-hydroxyvitamin D3 over a long period of time.


In vitro dissolution tests of the same formulations (dissolution media: 0.056 lipase in Ctab/NaH2PO4 buffer, pH 6.8) over a period of 120 minutes showed results generally consistent with the in vivo data (e.g., formulations 2 and 7 showed a more gradual and incomplete rise in % dissolution, whereas the immediate release control showed 100% release within 30 minutes).
























TABLE 2








25-
















(OH)-




Vitamin

Carnauba
GELUCIRE

Soybean

Hard


Liquid
MIGLYOL



Ingredient
D3
Ethanol
Wax
44/14
LABRASOL
Oil
BHT
Paraffin
GMS
GMO
Paraffin
812N
Total






























1
% w/w
0.12
2.32
14.63
9.75
9.75
63.40
0.02





100



mg/Cap
0.25
4.75
30.00
20.00
20.00
130.00
0.04





205


2
% w/w
0.12
2.32
27.50
9.75
9.75
50.53
0.02





100



mg/Cap
0.25
4.75
56.40
20.00
20.00
103.60
0.04





205


3
% w/w
0.12
2.32
14.63
9.75
37.85
35.31
0.02





100



mg/Cap
0.25
4.75
30.00
20.00
77.60
72.40
0.04





205


4
% w/w
0.12
2.32
11.51
8.10
3.12
74.80
0.02





100



mg/Cap
0.25
4.75
23.60
16.60
6.40
153.36
0.04





205


5
% w/w
0.12
2.32

9.75


0.02
14.63

37.85
35.31

100



mg/Cap
0.25
4.75

20.00


0.04
30.00

77.60
72.40

205


6
% w/w
0.12
2.32

9.75


0.02
14.63
9.75
9.75
53.65

100



mg/Cap
0.25
4.75

20.00


0.04
30.00
20.00
20.00
110.00

205


7
% w/w
0.12
2.32

9.75


0.02
27.51
37.85

22.43

100



mg/Cap
0.25
4.75

20.00


0.04
56.40
77.60

46.00

205


8
% w/w
0.12
2.32




0.02
9.75
9.75
9.75
68.23

100



mg/Cap
0.25
4.75




0.04
20.00
20.00
20.00
139.96

205


9
% w/w
0.12
2.32




0.02




97.54
100



mg/Cap
0.25
4.75




0.04




199.96
205









The data in Table 3 below show various pharmacokinetic parameters produced in the test subjects by administration of the Group 7 formulation according to the invention compared to the Group 9 prior art immediate release formulation and the Group 10 IV injection administration. The data demonstrate that the Group 7 formulation according to the invention avoided a concentration spike, provided a maximum concentration at a time much later than the immediate release dosage form and the intravenous injection, and provided a longer clearance half life than the comparable immediate release dosage form. The Group 7 formulation according to the invention resulted in a slower elimination of the 25-hydroxyvitamin D3 administered from systemic circulation compared to Group 9.


A single dose of 250 μg 25-hydroxyvitamin D3 administered according to the Group 7 formulation of the invention to mini-pigs (about 10 kg) resulted in an approximately 40 ng/ml rise in serum 25-hydroxyvitamin D3. A single dose of 50 μg 25-hydroxyvitamin D3 to a human (about 60 kg) is expected to increase serum levels of 25-hydroxyvitamin D3 by about 1.4 ng/ml.

















TABLE 3







AUC
AUC









(0-672 hr)
(0-INF)
Cmax
Tmax

Cmax24 hr/C24 hr
BA


Grp

(ng/ml hr)
(ng/ml hr)
(ng/ml)
(hr)
(hr)
(ng/ml)
(%)























7
AVG
8062.6
10425.7
39.5
39.2
120.9
1.42




STD
6259.2
6676.4
11.4
35.4
27.9
0.93
62.7



% RSD
77.63
64.0
28.7
90.2
23.0
65.41


9
AVG
12074.5
12201.4
204.8
3.5
71.5
2.23



STD
1028.0
1099.0
12.6
1.0
16.9
0.49
73.4



% RSD
8.5
9.0
6.1
28.6
23.7
22.11


10
AVG
15038.0
16616.1
154.9
1.5
132.4
2.12



STD
2903.4
3646.2
71.1
1.7
18.7
0.84
100.0



% RSD
19.3
21.9
45.9
112.0
14.1
39.67









Comparative Cmax, Tmax, and bioavailability data for the formulations of Groups 1-6 and 8 are shown in Table 4 below.













TABLE 4







Cmax
Tmax
BA


Group

(ng/ml)
(hr)
(%)



















1
AVG
105.9
7.0
69.1



STDEV
33.0
9.6



% RSD
31.2
137.0


2
AVG
29.7
12.8
25.3



STDEV
15.2
10.4



% RSD
51.2
80.9


3
AVG
109.4
4.0
84.1



STDEV
22.6
0.0



% RSD
20.6
0.0


4
AVG
162.1
4.8
97.2



STDEV
30.3
1.8



% RSD
18.7
37.3


5
AVG
90.8
3.2
70.7



STDEV
22.7
1.1



% RSD
24.9
34.2


6
AVG
99.9
3.2
72.3



STDEV
24.3
1.8



% RSD
24.4
55.9


8
AVG
91.5
3.6
70.2



STDEV
41.2
0.9



% RSD
45.0
24.8









Example 2—Pharmacokinetic Studies in Miniature Swine with Oral Capsules

The purpose of the study was to assess the systemic absorption of 25-hydroxyvitamin D3 in male Yucatan swine (about 45 kg body weight) following the administration of: a) 1×250 μg 25-hydroxyvitamin D3 modified release (MR) capsule, b) 2×250 μg MR capsules, c) 4×250 μg MR capsules, d) 1×1000 μg MR capsule, e) 1×250 μg immediate release (IR) 25-hydroxyvitamin D3 capsule, and f) 1×250 μg MR capsule administered on 3 consecutive days.


The MR formulations were prepared based on the formulation of Example 1, Group 7, above. In the case of the 1000 μg MR capsule, the higher concentration of 25-hydroxyvitamin D3 was offset by a relative decrease in ethanol.


To prepare the IR formulation 25-hydroxyvitamin D3 (0.12% wt/wt; 250 μg per capsule) was dissolved in ethanol USP (2.32% wt/wt; solublizer) and mixed with corn oil USP, (97.54% wt/wt; main vehicle) and butylated hydroxytoluene (0.02% wt/wt; antioxidant). The corn oil solution (205 mg) was filled into size 0 two piece hard gelatin capsules.


Eight male Yucatan miniature swine per group were each administered a dose based on the dosing schedule in Table 5 below. Blood was collected from animals prior to first dose and at 0.5, 1, 2, 4, 6, 8, 10, 12, 24, 48, 72, and 96 hrs following first dose. Animals in Group 6 were administered a second and third dose immediately following the collection of the 24 and 48 hr blood samples, respectively. 25-hydroxyvitamin D3 was assayed in all the collected samples. Ionized calcium and total calcium were determined in samples collected from animals in Group 1 and Group 5 at the following time points: pre dose and at 0.5, 1, 2, 4, 6, 8, 10, 12, 24, 48, 72, and 96 hrs following first dose.














TABLE 5








Number/





Group
Gender of
Dose



ID
Animals
Route
Dose/Animal









1M
8/male
Oral
1 capsule × 250 μg,






modified release



2M
8/male
Oral
2 capsules × 250 μg,






modified release



3M
8/male
Oral
4 capsules × 250 μg,






modified release



4M
8/male
Oral
1 capsule × 1000 μg,






modified release



5M
8/male
Oral
1 capsule × 250 μg,






immediate release



6M
8/male
Oral
3 capsules × 250 μg,






modified release










25-hydroxyvitamin D3 in swine serum was assayed using solid-phase extraction (SPE) with high performance liquid chromatography with tandem mass spectrometry (LC-MS/MS) detection. Serum samples were baseline corrected to exclude endogenous concentrations of 25-hydroxyvitamin D3 from the pharmacokinetic analysis. To achieve this pre-dose 25-hydroxyvitamin D3 concentration of each animal were subtracted from each of its post dose concentrations. Serum samples below the 1 ng/ml (lower limit of quantitation) were assigned a value of zero.


Pharmacokinetic parameters are reported in Table 6.

















TABLE 6







AUC(0-24 hr)
AUC(0-t)
Cmax
C24 hr
Tmax




Group

(ng/ml hr)
(ng/ml hr)
(ng/mL)
(ng/mL)
(hours)
Cmax/C24 hr
Cmax/AUC(0-24 hr)























1
AVG
417.81
1838.73
31.58
26.08
26.50
1.28
0.08



STDEV
121.63
709.85
7.63
9.87
22.42
0.28
0.02



% RSD
29.1
38.6
24.1
37.9
84.6
22.0
29.3


2
AVG
619.30
2862.75
47.86
36.80
30.50
1.42
0.10



STDEV
315.95
528.10
14.51
10.86
23.24
0.38
0.08



% RSD
51.0
18.4
30.3
29.5
76.2
26.4
79.4


3
AVG
1059.99
4321.75
72.29
58.00
25.50
1.28
0.07



STDEV
232.36
894.26
18.76
18.35
23.22
0.27
0.008



% RSD
21.9
20.7
26.0
31.6
91.1
21.1
11.5


4
AVG
642.79
2608.04
52.19
39.41
25.71
1.61
0.12



STDEV
392.48
1574.53
20.41
15.97
20.89
0.35
0.08



% RSD
61.1
60.4
39.1
40.5
81.3
21.5
67.2


5
AVG
812.51
2374.50
49.73
30.97
5.75
1.63
0.06



STDEV
115.47
266.95
9.22
4.76
1.28
0.34
0.005



% RSD
14.2
11.2
18.5
15.4
22.3
21.0
8.7









Dose normalized pharmacokinetic parameters for Groups 1 to 3 are reported in Table 7.













TABLE 7









Group 1
Group 2
Group 3
















PK Parameters
AVG
STDEV
% RSD
AVG
STDEV
% RSD
AVG
STDEV
% RSD



















AUC(0-t)
7.35
2.84
38.61
5.73
1.06
18.45
4.32
0.89
20.69


(ng/ml hr)/μg


Cmax (ng/mL)/μg
0.13
0.03
24.15
0.10
0.03
30.33
0.07
0.02
25.96


C24hr (ng/mL)/μg
0.10
0.04
37.85
0.07
0.02
29.50
0.06
0.02
31.64


AUC(0-24 h)
1.67
0.49
29.11
1.24
0.63
51.02
1.06
0.23
21.92


(ng/ml hr)/μg









For the groups administered 1, 2 and 4 capsules (250 μg MR capsules), there was an increase in exposure as a function of dose. Dose proportional exposure occurred at the 1×250 μg and 2×250 μg doses, while slightly less than proportional exposure was observed between 2×250 μg and 4×250 μg doses. The mean time at which maximum concentration was achieved (Tmax) was between 25.5 to 30.5 hrs.


Comparison of exposure from a single capsule (1×1000 μg) versus four capsules (4×250 μg) indicated higher exposure in animals dosed with multiple capsules. Dose independent parameters, such as mean Tmax, were similar for both dosing strategies.


The comparison of the modified release formulation of 25-hydroxyvitamin D3 (MR) (Group 1) to the IR formulation (Group 5), indicated that the MR formulation avoided a spike in the concentration of serum 25-hydroxyvitamin D3. The relative bioavailability of the MR formulation when compared to the IR formulation was approximately 77%. Animals receiving the MR formulation exhibited a mean Tmax of 26.5 hrs which indicated a significant delay compared to the animals receiving the IR formulation (Tmax=5.75 hrs).


Exposure was assessed in animals receiving 1×250 μg MR capsules on Days 1, 2 and 3. The mean increase in concentration of 25-hydroxyvitamin D3 over baseline 24 h following dosing was 17.3, 31.5 and 43.9 ng/mL following the first, second and third dose respectively.



FIG. 13 through FIG. 18 show the mean pharmacokinetic profile for animals in Groups 1-6, respectively. FIG. 19 shows a comparison of pharmacokinetic profiles for MR and JR formulations of 250 μg 25-hydroxyvitamin D3.


Example 3—Systemic Exposure Studies in Miniature Swine with Oral Capsules

The purpose of this study was to assess the increase in systemic 25-hydroxyvitamin D3 concentrations in healthy normal male Yucatan swine (˜50-60 kg body weight) maintained on a diet including an adequate intake of Vitamin D, following the daily administration of the following: a) 25 μg immediate release (IR) 25-hydroxyvitamin D3 capsules (Group 1), b) 25 μg modified release (MR) 25-hydroxyvitamin D3 capsules (Group 2), and c) 125 μg MR 25-hydroxyvitamin D3 capsules (Group 3), for 21 days.


The MR formulations were prepared based on the formulation of Example 1, Group 7, above. The differences in concentration of 25-hydroxyvitamin D3 were offset by relative changes in ethanol.


To prepare the IR formulation 25-hydroxyvitamin D3 (0.12% wt/wt; 250 μg per capsule) was dissolved in ethanol USP (2.32% wt/wt; solublizer) and mixed with corn oil USP, (97.54% wt/wt; main vehicle) and butylated hydroxytoluene (0.02% wt/wt; antioxidant). The corn oil solution (205 mg) was filled into size 0 two piece hard gelatin capsules.


Eight male Yucatan miniature swine per group were each administered a daily dose based on the dosing schedule in Table 8, below.












TABLE 8






Number/




Group
Gender of
Dose


ID
Animals
Route
Dose/Animal







1M
8/male
Oral
1 × 25 μg, immediate release 25-





hydroxyvitamin D3 capsule dosed daily





for 21 days


2M
8/male
Oral
1 × 25 μg, modified release 25-





hydroxyvitamin D3 capsule dosed daily





for 21 days


3M
8/male
Oral
1 × 125 μg, modified release 25-





hydroxyvitamin D3 capsule dosed daily





for 21 days









Blood was collected from animals prior to the first dose and daily at 24 h following daily dose, prior to subsequent dose. The concentration of serum 25-hydroxyvitamin D3 was assayed using solid-phase extraction (SPE) with high performance liquid chromatography with tandem mass spectrometry (LC-MS/MS) detection. Total serum calcium was determined in samples collected from animals at the following time points: pre dose (Day 0) and 24 h following last dose (Day 21).


In all three groups, pre-dose mean serum 25-hydroxyvitamin D3 concentrations were approximately 26 ng/mL. After 21 doses, an increase in serum 25-hydroxyvitamin D3 was observed in all animals. Following the repeat administration of 25 μg MR or IR capsules, the concentration of serum 25-hydroxyvitamin D3 increased to levels above 30 ng/mL and began to plateau at approximately 45 and 55 ng/mL, respectively at approximately Day 17 to 18. Upon the administration of a single dose, the increase in serum 25-hydroxyvitamin D3 between the two regimens was comparable (3.84 versus 4.18 ng/mL). On the other hand, at the completion of dosing the increase was approximately 60% greater for animals administered the IR formulation. This finding indicates that the bioavailability from the MR capsule is comparable to that of the IR following a single dose, but that the MR capsules present a method for repeat dosing of 25-hydroxyvitamin D3 in which systemic 25-hydroxyvitamin D3 can be gradually increased.


Animals administered 125 μg MR capsules exhibited higher levels of serum 25-hydroxyvitamin D3. The administration of a 5 fold greater dose (125 μg versus 25 μg MR capsules) resulted in approximately 5 fold greater increase in 25-hydroxyvitamin D3 following single and repeated dose. This finding indicates that exposure from MR capsules is dose proportional from 25 to 125 μg.


The effect of the administration of IR and MR capsules on the concentration of serum calcium was investigated. After the administration of 21 doses of either IR or MR, the levels of calcium in serum did not change from pre-dose baseline levels. This finding indicates that 25-hydroxyvitamin D3 MR capsules can be utilized to increase serum 25-hydroxyvitamin D3 levels to above 100 ng/mL without causing an increase in serum calcium.


Mean uncorrected serum 25-hydroxyvitamin D3 concentration versus time profiles for Groups 1 to 3 are illustrated in FIG. 20. Mean baseline corrected serum 25-hydroxyvitamin D3 concentration versus time profiles for Groups 1 to 3 are illustrated in FIG. 21, FIG. 22, and FIG. 23, respectively.



FIG. 24 shows the mean change in parathyroid hormone levels for Group 1 animals from predose to day 21, and FIG. 25 shows the mean change in parathyroid hormone levels for Group 2 animals from predose to day 21. The immediate release and modified release formulations as administered in this study both raised serum 25-hydroxyvitamin D3; however, the immediate release formulation resulted in undesirable pharmacological decreases in PTH in these subjects. The modified release formulation as administered did not effect acute supraphysiological reductions in PTH and allows for gradual PTH lowering, believed to be associated with physiological adaptation to markedly rising 25-hydroxyvitamin D3 levels. The MR formulation should permit attainment of higher serum 25-hydroxyvitamin D3 levels without safety concerns associated with undesirable pharmacological lowering of PTH.


Example 4—Pharmacokinetic Studies in Beagle Dogs with Oral Capsules

Modified release 25-hydroxyvitamin D3 capsules were administered daily to Beagle dogs (10 kg) for 13 consecutive weeks. The MR formulations were prepared based on the formulation of Example 1, Group 7, above. The differences in concentration of 25-hydroxyvitamin D3 were offset by relative changes in ethanol.


The capsules were administered orally, as shown in Table 9 below.












TABLE 9






Nominal Dose Level




Treatment
(μg/kg/day), based
Dose/Capsule
Number


Group
on 10 kg ave. weight
(μg)
of Capsules


















1. Control
0
0
1


Group (placebo)


2. Low Dose
2.5
25
1


3. Mid-Low
12.5
125
1


Dose


4. Mid-High
50
500
1


Dose


5. High Dose
100
1000
1









Dogs were bled prior to the first dose and at specific time points following the first dose, up to 13 weeks (92 days). Serum was generated and 25-hydroxyvitamin D3 was assayed in the serum using a liquid chromatography tandem mass spectrometry method.


Mean serum 25-hydroxyvitamin D3 concentration versus time profiles for Groups 1 to 5 are illustrated in FIG. 26.


Example 5—Release Upon Dissolution


FIG. 27 shows a dissolution release profile for 250 μg capsules according to Example 2 above, which showed an average release of about 72% of 25-hydroxyvitamin D3 at 24 hours. As described above, preferably the modified release formulation releases about 80% of the drug in the first 24 hours.


Example 6—Efficacy Study in Healthy Adult Male Volunteers with Vitamin D Insufficiency

The effectiveness of three different formulations of Vitamin D in restoring serum 25-hydroxyvitamin D to optimal levels (>30 ng/mL) is examined in a 23-day study of healthy non-obese men diagnosed with Vitamin D insufficiency. One of the formulations (Formulation #1) is a soft gelatin capsule containing 30 μg of 25-hydroxyvitamin D3 prepared as described in Example 1, Group 7, above. The second formulation (Formulation #2) is an immediate-release soft gelatin capsule of identical appearance containing 50,000 IU of ergocalciferol dissolved in medium chain triglyceride oil. The third formulation (Formulation #3) is an immediate-release soft gelatin capsule, also of identical appearance, containing 50,000 IU of cholecalciferol dissolved in medium chain triglyceride oil. A total of 100 healthy Caucasian and African-American men participate in this study, all of whom are aged 30 to 45 years and have serum 25-hydroxyvitamin D levels between 15 and 29 ng/mL (inclusive). All subjects abstain from taking other Vitamin D supplements for 60 days before study start and continuing through study termination, and from significant sun exposure. On Day 1 and 2 of the study, all subjects provide fasting morning blood samples to establish pre-treatment baseline values of serum 25-hydroxyvitamin D. On the morning of Day 3, the subjects provide an additional fasting blood sample (t=0), are randomly assigned to one of four treatment groups, and are dosed with a single test capsule prior to eating breakfast: the subjects in Group #1 each receive a single capsule of Formulation #1, and the subjects in Groups #2 and #3 each receive a single capsule of Formulation #2 or Formulation #3, respectively. Subjects in Group #4 receive a matching placebo capsule. Subjects in Group #1 each receive an additional capsule of Formulation #1 on the mornings of Days 4 through 22 before breakfast, but subjects in Groups #2, #3 and #4 receive no additional capsules. A fasting morning blood sample is drawn from each subject, irrespective of treatment group, on Days 4, 5, 6, 10, 17 and 23 (or 1, 2, 3, 7, 14 and 20 days after the start of dosing). All collected blood is analyzed for the contained levels of 25-hydroxyvitamin D and PTH, and the data are analyzed by treatment group after correction for baseline values. Subjects in all four treatment groups exhibit mean baseline serum 25-hydroxyvitamin D levels of approximately 16 to 18 ng/mL, based on analysis of fasting blood samples drawn on Days 1 through 3. Subjects in Group #4 (control group) show no significant changes in mean serum 25-hydroxyvitamin D over the course of the study. Subjects in Group #1 show a steadily increasing mean serum 25-hydroxyvitamin D reaching at least 30 ng/mL by Day 23 and decreases in plasma PTH. In marked contrast, subjects in Group #2 exhibit marked increases in mean serum 25-hydroxyvitamin D for the first few days post-dosing, reaching a maximum of 29 ng/ml and then rapidly declining thereafter. By study end, serum 25-hydroxyvitamin D is significantly lower than baseline in Group #2. and plasma PTH is not decreased. Subjects in Group #3 exhibit continuing increases in mean serum 25-hydroxyvitamin D through the first 2 weeks after dosing with gradual, but progressive, decreases occurring thereafter. By study end, mean serum 25-hydroxyvitamin D is below 30 ng/mL, being only approximately 11 ng/mL higher than pre-treatment baseline and plasma PTH is only marginally decreased. The data from this study demonstrate that administration of 600 μg of 25-hydroxyvitamin D3, formulated as described herein and administered at a dose of 30 μg per day for 20 days, is substantially more effective in restoring low serum levels of 25-hydroxyvitamin D to optimal levels than immediate-release formulations of 50,000 IU of either ergocalciferol or cholecalciferol administered in single doses, as currently recommended by the NKF and other leading experts on oral Vitamin D replacement therapy.


Example 7—Efficacy Study in Patients with Stage 4 CKD and Secondary Hyperparathyroidism Associated with Vitamin D Insufficiency

The effectiveness of oral immediate-release and modified-release 25-hydroxyvitamin D3 in restoring serum total 25-hydroxyvitamin D to optimal levels (>30 ng/mL) is examined in a 6-month study of adult male and female patients with Stage 4 CKD and secondary hyperparathyroidism associated with vitamin D insufficiency. Two formulations are used in the study. One of the formulations (Formulation #1) is a soft gelatin capsule containing 40 μg of 25-hydroxyvitamin D3 in a modified-release formulation. The second formulation (Formulation #2) is a soft gelatin capsule containing 40 μg of 25-hydroxyvitamin D3 in an immediate-release formulation. A total of 100 subjects participate in this study, all of whom are aged 30 to 70 years and have serum 25-hydroxyvitamin D levels between 15 and 29 ng/mL (inclusive) and serum intact parathyroid hormone (iPTH) levels above the target levels published in the current K/DOQI Guidelines at the time of enrolment. All subjects abstain from taking other Vitamin D supplements for 60 days before study start and continuing through study termination, and from significant sum exposure. All subjects begin daily dosing with two capsules of either Formulation #1 or Formulation #2. Serum total 25-hydroxyvitamin D is measured at biweekly intervals and serum iPTH is determined at quarterly intervals. After 1 month, the daily dosage of both Formulations is maintained unchanged in patients whose serum total 25-hydroxyvitamin D is between 50 and 90 ng/mL, increased by one capsule in patients whose serum total 25-hydroxyvitamin D is below 50 ng/mL, and decreased by one capsule per day in patients whose serum total 25-hydroxyvitamin D is above 90 ng/mL. Further adjustments in the daily dose are made in order to maintain serum total 25-hydroxyvitamin D between 50 and 90 ng/mL. Dosing with both Formulation #1 and #2 is continued indefinitely, provided that hypercalcemia, hypercalciuria and hyperphosphatemia do not develop, in which case appropriate adjustments in dosage are made. After 6-months, the subjects' serum total 25-hydroxyvitamin D levels are found to remain stable between 50 and 90 ng/mL with treatment with Formulation #1 and serum iPTH is found to remain stable at levels consistent with or closer to targets published in the K/DOQI Guidelines. The incidence of hypercalcemia, hypercalciuria and hyperphosphatemia are rare once stable dosing has been achieved. In contrast after 6-months, the subjects' serum total 25-hydroxyvitamin D levels are not found to remain stable between 50 and 90 ng/mL with treatment with Formulation #2 and serum iPTH does not reach levels consistent with or closer to targets published in the K/DOQI Guidelines. The incidence of hypercalcemia, hypercalciuria and hyperphosphatemia are substantial.


Data from this study demonstrate that the modified release formulation of 25-hydroxyvitamin D3 is effective at increasing serum 25-hydroxyvitamin D without causing unacceptable side effects related to calcium and PTH metabolism.


Example 8—Pharmacodynamic and Pharmacokinetic Profiles of Modified Release Capsules and Intravenous Calcifediol for Secondary Hyperparathyroidism in Stage 3 or 4 CKD

In this randomized open label single-dose study, 27 subjects with baseline serum total 25-hydroxyvitamin D between 16 and 29 ng/mL and serum iPTH above K/DOQI targets were dosed with a modified release oral dosage form according to the disclosure herein (450 or 900 μg) or IV calcifediol (25-hydroxyvitamin D3) (448 μg) to evaluate bioavailability, pharmacokinetics, pharmacodynamics, safety and tolerability. The modified release dosage formulation was the same as formulation 7 in Table 2 herein, with the following exceptions: (1) a vegetable-based capsule was used in place of a gelatin capsule; (2) hard paraffin content was reduced to 20%; and (3) liquid paraffin (light mineral oil) content was increased to 30%. The amount of 25-hydroxyvitamin D3 per capsule was 90 μg. The bioavailability was estimated to be about 7% measured over the first 24 hours post-dose and about 10% over the total 42-day follow-up period.


Serum calcium (Ca), phosphorus (P), iPTH, 25-hydroxyvitamin D3 and total 1,25-dihydryoxyvitamin D were monitored for 6 weeks. Mean (±SD) baseline 25-hydroxyvitamin D3 and iPTH were 20±10 ng/mL and 197±146 pg/mL, respectively. IV calcifediol rapidly raised mean 25-hydroxyvitamin D3 to 134±19 ng/mL. The modified release capsules produced gradual increases in 25-hydroxyvitamin D3 to a mean of 25±10 ng/mL (450 μg) and 32±16 ng/mL (900 μg). Serum 25-hydroxyvitamin D3 Tmax for the IV calcifediol was 0.5±0.6 hours, whereas Tmax for the modified release dosage forms was 13.1±9.6 hours (450 μg) and 13.6±10 hours (900 μg). The bioavailability of the modified release dosage form was about 10% measured over the duration of the study (42 days). No confirmed hypercalcemia (>10.3 mg/dL) was observed in any treatment group. Serum total 1,25-dihydroxyvitamin D reached an increase of 18 pg/mL at 90 hrs after IV calcifediol, and of 15 and 9 pg/mL at 100 to 200 hrs after 450 and 900 μg of the modified release capsules, respectively. IV calcifediol and 450 μg of the modified release capsules had no clinically meaningful effect on mean iPTH. The modified release capsules dosed at 900 μg reduced iPTH from baseline by a mean of 19% at 24 hrs. The rapid increase in serum total 1,25-dihydroxyvitamin D after IV calcifediol may have triggered excessive expression of the vitamin D catabolic enzyme, CYP24, in the parathyroid glands, leading to local hormone resistance and limited iPTH suppression.


The elimination half-life (t1/2) of the absolute 25-hydroxyvitamin D3 levels were substantially increased following administration of the modified release capsules (2478 and 3229 hours) compared to that following IV calcifediol (1776 hours). The elimination half-life (t1/2) of the absolute 1,25-hydroxyvitamin D levels also were substantially increased following administration of the modified release capsules (3975 and 3833 hours) compared to that following IV calcifediol (2216 hours).


Table 10 and Table 11 show a summary of the observed and baseline-adjusted pharmacokinetic parameters, respectively, for 25-hydroxyvitamin D3 by treatment group. Table 12 and Table 13 show a summary of the observed and baseline-adjusted pharmacokinetic parameters, respectively, for 1,25-dihydroxyvitamin D3 by treatment group.











TABLE 10







PK
450 μg MR (N = 9)
900 μg MR (N = 9)













Parameters
AVG
STDEV
Median
Range
AVG
STDEV





AUC(0-42 days)
13353.12
10606.47
12196.45
888.39,
21563.95
9165.53


(ng * h/ml)



32885.95


AUC(0-last)
13353.12
10606.47
12196.45
888.39,
21563.95
9165.53


(ng * h/ml)



32885.95


AUC(0-inf)
81511.71
103037.08
54967.57
4927.95,
122901.73
114168.13


(ng * h/ml)



333366.90


Cmax
25.18
10.134
20.52
15.35,
31.54
15.765


(ng/mL)



42.24


Clast (ng/mL)
18.11
7.846
15.84
10.30,
19.08
7.611






29.80


tmax (h)
13.11
9.597
10.00
6.00,
13.56
9.989






36.00


λz (h−1)
0.0015
0.0028
0.0003
0.0001,
0.0003
0.0002






0.0087


R2
0.89
0.130
0.96
0.662,
0.90
0.169






1.000


t1/2 (h)
2477.72
2581.24
2483.61
79.24,
3228.63
2734.74






8615.11


Vd (L/ng)
49.42
18.30
50.93
23.20,
45.06
19.38






72.76


CL (L/ng * h)
0.0499
0.0155
0.0182
0.0030,
0.0155
0.0119






0.2029


F
0.306
(NA)
NA
NA
0.247
NA












PK
900 μg MR (N = 9)
450 μg IV (N = 9)













Parameters
Median
Range
AVG
STDEV
Median
Range





AUC(0-42 days)
17940.11
9292.88,
43463.51
12589.57
47115.66
23340.59,


(ng * h/ml)

38631.91



63006.78


AUC(0-last)
17940.11
9292.88,
43463.51
12589.57
47115.66
23340.59,


(ng * h/ml)

38631.91



63006.78


AUC(0-inf)
79902.04
25729.84,
137955.58
66746.71
123580.87
39282.49,


(ng * h/ml)

378935.59



243322.76


Cmax
30.12
12.21,
133.99
19.311
133.68
91.71,


(ng/mL)

67.01



160.91


Clast (ng/mL)
21.37
7.30,
35.07
12.330
36.91
12.68,




27.93



53.39


tmax (h)
10.00
2.00,
0.49
0.638
0.25
0.083,




30.00



2.00


λz (h−1)
0.0004
0.0001,
0.0005
0.0002
0.0004
0.0002,




0.0005



0.0008


R2
0.99
0.523,
0.91
0.090
0.93
0.730,




0.998



1.000


t1/2 (h)
1937.32
1300.13,
1775.86
779.13
1694.69
871.46,




9646.71



3297.78


Vd (L/ng)
40.080
20.77,
20.35
7.42
19.550
13.04,




87.51



32.68


CL (L/ng * h)
0.0125
0.0026,
0.0095
0.0065
0.0081
1.000,




0.0389



NA


F
NA
NA
1.000
NA
NA
NA


















TABLE 11







PK
450 μg MR (N = 9)
900 μg MR (N = 9)














Parameters
AVG
STDEV
Median
Range
AVG
STDEV
Median





AUC(0-42 days)
1394.89
1911.41
605.42
48.87,
4525.43
3123.29
4801.54


(ng * h/ml)



4956.68


AUC(0-last)
1257.30
2047.74
48.87
−325.43,
4274.27
3488.40
4801.54


(ng * h/ml)



4956.68


AUC(0-inf)
3318.90
4606.89
547.25
50.78,
6791.49
5224.54
6872.86


(ng * h/ml)



9878.03


Cmax
6.90
4.266
5.70
2.93,
14.17
9.884
12.30


(ng/mL)



14.87


Clast
2.36
2.257
2.05
0.16,
2.64
1.784
3.01


(ng/mL)



5.49


tmax (h)
13.11
9.597
10.00
6.00,
15.00
9.621
10.00






36.00


λz (h−1)
0.018
0.359
0.0042
0.0008,
0.0020
0.0013
0.0016






0.0910


R2
0.92
0.101
0.96
0.743,
0.99
0.020
0.99






0.999


t1/2 (h)
307.86
336.42
165.72
7.61,
522.96
320.80
530.00






914.23


Vd (L/ng)
340.42
269.11
249.77
80.51,
82.92
29.70
77.11






771.79


CL (L/ng * h)
4.588
7.525
2.119
0.101,
0.141
0.080
0.1200






19.692


F
0.064
N/A
N/A
N/A
0.109
N/A
N/A














PK
900 μg MR (N = 9)
450 μg IV (N = 9)














Parameters
Range
AVG
STDEV
Median
Range







AUC(0-42 days)
148.07,
19609.07
5319.01
18764.48
11066.02,



(ng * h/ml)
8843.74



28611.05



AUC(0-last)
−877.99,
19609.07
5319.01
18764.48
11066.02,



(ng * h/ml)
8843.74



28611.05



AUC(0-inf)
285.04,
34543.75
22103.97
26962.25
16868.97,



(ng * h/ml)
14979.17



89126.53



Cmax
2.55,
110.33
14.536
111.08
76.63,



(ng/mL)
35.59



127.40



Clast
0.42,
11.40
5.648
9.01
5.84,



(ng/mL)
5.40



22.86



tmax (h)
8.00,
0.49
0.638
0.25
0.083,




30.00



2.00



λz (h−1)
0.0008,
0.0011
0.0005
0.0010
0.0004,




0.0037



0.0021



R2
0.948,
0.92
0.086
0.95
0.720,




1.000



0.999



t1/2 (h)
189.84,
745.86
437.65
663.43
337.37,




879.31



1834.71



Vd (L/ng)
39.96,
32.94
11.00
29.70
24.28,




127.06



58.95



CL (L/ng * h)
0.0668,
0.036
0.014
0.037
0.011,




0.2860



0.059



F
N/A
1.000
N/A
N/A
N/A




















TABLE 12







PK
450 μg MR (N = 9)
900 μg MR (N = 9)
450 μg IV (N = 9)
















Parameters
AVG
Median
Range
AVG
Median
Range
AVG
Median
Range



















AUC(0-last)
21364.61
21904.55
14508.53,
29202.83
28607.65
16659.23,
26943.27
28193.71
11392.60,


(pg * h/ml)


30551.35


59589.95


47683.28


Cmax (pg/mL)
30.54
29.30
17.6,
41.12
39.90
25.4, 88.5
37.27
41.80
15.7, 57.2





52.2


Clast (pg/mL)
20.00
20.50
11.7,
26.57
22.50
15.9, 59.5
24.39
29.70
7.9, 37.3





31.1


tmax (h)
152.0
36.0
2, 672
193.3
168.0
6, 504
93.3
48.0
4, 336


R2
0.45
0.44
0.01,
0.41
0.26
0.08, 0.98
0.65
0.82
0.05,





0.94





1.00


t1/2 (h)
3974.82
2513.67
1249.92,
3833.14
2318.35
984.87,
2216.12
1799.15
687.02,





13893.01


10210.62


5743.86



















TABLE 13







PK
450 μg MR (N = 9)
900 μg MR (N = 9)
450 μg IV (N = 9)
















Parameters
AVG
Median
Range
AVG
Median
Range
AVG
Median
Range



















AUC(0-last)
−4.99
−435.07
−3173.48,
2530.03
3591.45
−9444.35,
7448.74
6803.18
2355.31,


(pg * h/ml)


4025.08


14784.35


19364.96


Cmax
9.34
6.53
3.08,
14.66
10.68
5.05, 44.05
17.93
17.98
7.20, 35.83


(pg/mL)


20.05


Clast
−1.2
−0.73
−5.08, 1.48
0.11
0.07
−10.05,
5.05
4.07
−1.93, 14.93


(pg/mL)





15.05


tmax (h)
152.0
36.0
2, 672
193.3
168.0
6, 504
93.3
48.0
4, 336


R2
0.45
0.44
0.01, 0.94
0.41
0.26
0.08, 0.98
0.65
0.82
0.05, 1.00


t1/2 (h)
3974.82
2513.67
1249.92,
3833.14
2318.35
984.87,
2216.12
1799.15
687.02,





13893.01


10210.62


5743.86









These findings demonstrate that the modified release capsules gradually normalized 25-hydroxyvitamin D3 levels and suppressed elevated iPTH in this patient population, and that the mechanism to lower iPTH may be more complex than simply boosting serum total 25-hydroxyvitamin D.



FIG. 28 shows a plot of the change in serum 25-hydroxyvitamin D3 levels over 96 hours in Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were intravenously (N=8) or orally (N=8) administered a single dose of 450 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein or orally administered (N=8) a single dose of 900 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein. The orally administered doses of 25-hydroxyvitamin D3 gradually increased levels of serum 25-hydroxyvitamin D3, sustained this increase for over 96 hours, and avoided a surge in 25-hydroxyvitamin D3 serum levels. The orally administered dose having 900 μg of 25-hydroxyvitamin D3 increased levels of serum 25-hydroxyvitamin D3 by approximately 10 ng/ml at 96 hours versus only 5 ng/ml using the dose having 450 μg of 25-hydroxyvitamin D3.



FIG. 29 shows a plot of the change in serum 1,25-hydroxyvitamin D3 levels over 96 hours in Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were intravenously (N=8) or orally (N=8) administered a single dose of 450 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein or orally administered (N=8) a single dose of 900 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein. The orally administered doses of 25-hydroxyvitamin D3 gradually increased levels of serum 1,25-hydroxyvitamin D3, sustained this increase for over 96 hours, and avoided a surge in 1,25-hydroxyvitamin D3 serum levels. The orally administered dose having 900 μg of 25-hydroxyvitamin D3 increased levels of serum 1,25-hydroxyvitamin D3 to a greater extent than the orally administered dose having 450 μg of 25-hydroxyvitamin D3, which increased 1,25-hydroxyvitamin D3 by only about 2.5 pg/ml.



FIG. 30 shows the mean percent change in parathyroid hormone levels for Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were orally administered a single dose of 900 μg of 25-hydroxyvitamin D3 in a modified release dosage form as described herein (N=8) compared to subjects intravenously administered a single dose of 448 μg of 25-hydroxyvitamin D3 (N=8). The latter group served as a control (100% of baseline).



FIG. 31 shows the mean % change in iPTH levels for Stage 3 and 4 Chronic Kidney Disease subjects with vitamin D insufficiency and secondary hyperparathyroidism that were intravenously administered a single dose of 448 μg of 25-hydroxyvitamin D3 (N=8) or orally administered a single dose of 450 μg (N=8) or 900 μg (N=8) of 25-hydroxyvitamin D3 in a modified release dosage form. The orally administered single dose of 900 μg produced a steady decline with a sustained reduction in iPTH with a final mean % reduction of 16.8%.


The serum calcium levels remained within the normal range throughout the studies demonstrated in FIG. 28 through FIG. 31. In addition, no serious adverse events were reported.


The foregoing description is given for clearness of understanding only, and no unnecessary limitations should be understood therefrom, as modifications within the scope of the invention may be apparent to those having ordinary skill in the art.


Throughout the specification, where compositions are described as including components or materials, it is contemplated that the compositions can also consist essentially of, or consist of, any combination of the recited components or materials, unless described otherwise.


The practice of a method disclosed herein, and individual steps thereof, can be performed manually and/or with the aid of electronic equipment. Although processes have been described with reference to particular embodiments, a person of ordinary skill in the art will readily appreciate that other ways of performing the acts associated with the methods may be used. For example, the order of various of the steps may be changed without departing from the scope or spirit of the method, unless described otherwise. In addition, some of the individual steps can be combined, omitted, or further subdivided into additional steps.

Claims
  • 1. A method of treating secondary hyperparathyroidism, comprising orally administering to an adult human patient suffering from secondary hyperparathyroidism a sustained release dosage form comprising 25-hydroxyvitamin D3, in a dosage amount of 900 μg per week, providing a rise in serum total 25-hydroxyvitamin D of at least 7 ng/ml and no greater than 30 ng/ml within the first 24 hours after said administering, and said administering providing a rise in serum total 1,25-dihydroxyvitamin D of greater than 3 pg/mL and less than 10 pg/mL within the first 24 hours after said administering, wherein the rise in serum total 1,25-dihydroxyvitamin D is sustained for at least 48 hours following said administering, and lowering the patient's serum intact parathyroid hormone (iPTH) level.
  • 2. The method of claim 1, wherein the Tmax of serum total 25-hydroxyvitamin D following said administering step is at least 4 hours.
  • 3. The method of claim 2, wherein the Tmax of serum total 25-hydroxyvitamin D following said administering step is at least 12 hours.
  • 4. The method of claim 1, wherein the patient also suffers from vitamin D insufficiency or deficiency, as defined by serum total 25-hydroxyvitamin D of less than 30 ng/ml.
  • 5. The method of claim 1, comprising administering the sustained release dosage form on a frequency of every other day or less.
  • 6. The method of claim 1, wherein the patient's iPTH level is lowered by at least 10% compared to baseline.
  • 7. The method of claim 6, wherein the patient's iPTH level is lowered by at least 15% compared to baseline.
  • 8. The method of claim 1, wherein the patient's iPTH level remains lowered for at least 96 hours.
  • 9. The method of claim 1, wherein the patient suffers from hyperparathyroidism secondary to chronic kidney disease (CKD).
  • 10. The method of claim 9, wherein said CKD is Stage 2, Stage 3, Stage 4, or Stage 5 CKD.
  • 11. The method of claim 10, wherein said CKD is Stage 3 or Stage 4.
CROSS-REFERENCE TO RELATED APPLICATIONS

The benefit under 35 U.S.C. § 119(e) of U.S. Provisional Patent Application Ser. Nos. 61/318,741 filed Mar. 29, 2010, and 61/405,196 filed Oct. 20, 2010, is hereby claimed, and the disclosures thereof are hereby incorporated by reference herein.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US2011/030404 3/29/2011 WO 2/12/2013
Publishing Document Publishing Date Country Kind
WO2011/123476 10/6/2011 WO A
US Referenced Citations (241)
Number Name Date Kind
3565924 DeLuca et al. Feb 1971 A
3833622 Babcock et al. Sep 1974 A
3880894 De Luca et al. Apr 1975 A
3974272 Polli et al. Aug 1976 A
4004003 Babcock et al. Jan 1977 A
4230701 Holick et al. Oct 1980 A
4335120 Holick et al. Jun 1982 A
4442093 Maeda et al. Apr 1984 A
4448721 DeLuca et al. May 1984 A
4555364 DeLuca et al. Nov 1985 A
4668517 Weber et al. May 1987 A
4684524 Eckenhoff et al. Aug 1987 A
4695591 Hanna et al. Sep 1987 A
4721613 Urquhart et al. Jan 1988 A
4729895 Makino et al. Mar 1988 A
4755544 Makino et al. Jul 1988 A
4892821 Omura et al. Jan 1990 A
4997824 Popovtzer et al. Mar 1991 A
5026559 Eichel et al. Jun 1991 A
5160742 Mazer et al. Nov 1992 A
5167965 Schulz Dec 1992 A
5328903 Ishii et al. Jul 1994 A
5342626 Winston, Jr. et al. Aug 1994 A
5354743 Thys-Jacobs Oct 1994 A
5403831 DeLuca et al. Apr 1995 A
5431917 Yamamoto et al. Jul 1995 A
5472712 Oshlack et al. Dec 1995 A
5487900 Itoh et al. Jan 1996 A
5529991 Knutson et al. Jun 1996 A
5593690 Akiyama et al. Jan 1997 A
5602116 Knutson et al. Feb 1997 A
5614513 Knutson et al. Mar 1997 A
5622941 Knutson et al. Apr 1997 A
5693615 Stone Dec 1997 A
5707980 Knutson et al. Jan 1998 A
5756123 Yamamoto et al. May 1998 A
5783215 Arwidsson et al. Jul 1998 A
5795882 Bishop et al. Aug 1998 A
5861386 Knutson et al. Jan 1999 A
5869473 Knutson et al. Feb 1999 A
5872113 Nestor et al. Feb 1999 A
5888994 Hennessy et al. Mar 1999 A
5919986 Barbier et al. Jul 1999 A
5939408 Batcho et al. Aug 1999 A
5958451 Chen Sep 1999 A
5976784 DeLuca et al. Nov 1999 A
6001884 Nemeth et al. Dec 1999 A
6011068 Nemeth et al. Jan 2000 A
6031003 Nemeth et al. Feb 2000 A
6034075 Thys-Jacobs Mar 2000 A
6051567 Abrahamson et al. Apr 2000 A
6096876 St-Arnaud et al. Aug 2000 A
6121469 Norman et al. Sep 2000 A
6133250 Knutson et al. Oct 2000 A
6139875 Adams et al. Oct 2000 A
6147064 Knutson et al. Nov 2000 A
6150346 Knutson et al. Nov 2000 A
6190591 van Lengerich Feb 2001 B1
6190695 Hoshino et al. Feb 2001 B1
6211244 Van et al. Apr 2001 B1
6214376 Gennadios Apr 2001 B1
6228849 Thys-Jacobs May 2001 B1
6242434 Bishop et al. Jun 2001 B1
6265392 Abrahamson et al. Jul 2001 B1
6274169 Abrahamson et al. Aug 2001 B1
6288849 Teramoto Sep 2001 B1
6313146 Van et al. Nov 2001 B1
6340473 Tanner et al. Jan 2002 B1
6342249 Wong et al. Jan 2002 B1
6375981 Gilleland et al. Apr 2002 B1
6376479 Knutson et al. Apr 2002 B1
6380408 Posner et al. Apr 2002 B1
6413463 Yamamoto et al. Jul 2002 B1
6432936 DeLuca et al. Aug 2002 B1
6491950 Gutierrez-Rocca et al. Dec 2002 B1
6503893 Bishop et al. Jan 2003 B2
6521608 Henner et al. Feb 2003 B1
6524788 Cantor Feb 2003 B1
6569463 Patel et al. May 2003 B2
6572888 Byrd Jun 2003 B2
6582727 Tanner et al. Jun 2003 B2
6596314 Wong et al. Jul 2003 B2
6627622 DeLuca et al. Sep 2003 B2
6645527 Oshlack et al. Nov 2003 B2
6770295 Kreilgård et al. Aug 2004 B1
6887493 Shefer et al. May 2005 B2
6893658 Iida et al. May 2005 B1
6903083 Knutson et al. Jun 2005 B2
6911217 Gren et al. Jun 2005 B1
6923988 Patel et al. Aug 2005 B2
6929803 Wong et al. Aug 2005 B2
6949256 Fonkwe et al. Sep 2005 B2
6982258 Posner et al. Jan 2006 B2
RE39079 Tanner et al. Apr 2006 E
7033996 Christakos Apr 2006 B2
7056655 Cantor Jun 2006 B2
7101865 Posner et al. Sep 2006 B2
7122530 Bishop et al. Oct 2006 B2
7166585 Posner et al. Jan 2007 B2
7189843 Tsai et al. Mar 2007 B2
7226932 Gokhale et al. Jun 2007 B2
7255921 Kamaguchi et al. Aug 2007 B2
7422758 Block et al. Sep 2008 B2
7528122 DeLuca et al. May 2009 B2
7632518 Tritsch et al. Dec 2009 B2
7648826 Albertson et al. Jan 2010 B1
7807194 Modliszewski et al. Oct 2010 B2
7816341 Sewall et al. Oct 2010 B2
7829595 Lawrence et al. Nov 2010 B2
7846475 Shiraishi et al. Dec 2010 B2
7973024 Posner et al. Jul 2011 B2
8088410 Tritsch et al. Jan 2012 B2
8101203 Cao Jan 2012 B2
8101204 Cao Jan 2012 B2
8142811 Oshlack et al. Mar 2012 B2
8207149 Tabash Jun 2012 B2
8231896 Tanner et al. Jul 2012 B2
8268352 Vaya et al. Sep 2012 B2
8293270 Sukuru Oct 2012 B2
8329677 Bishop Dec 2012 B2
8361488 Bishop Jan 2013 B2
8377470 Tanner et al. Feb 2013 B2
8426391 Bishop Apr 2013 B2
8592401 Petkovich et al. Nov 2013 B2
8759328 Deluca Jun 2014 B2
8778373 Bishop Jul 2014 B2
8906410 Bishop et al. Dec 2014 B2
8962239 Petkovich Feb 2015 B2
8992971 Yang Mar 2015 B2
9017720 Andersen et al. Apr 2015 B2
9125823 Selva et al. Sep 2015 B2
9402855 Bishop et al. Aug 2016 B2
9408858 Bishop et al. Aug 2016 B2
9498486 Bishop Nov 2016 B1
9500661 Petkovich et al. Nov 2016 B2
9913852 Bishop et al. Mar 2018 B2
9918940 Bishop Mar 2018 B2
10220047 Petkovich et al. Mar 2019 B2
20010028896 Byrd Oct 2001 A1
20010036472 Wong et al. Nov 2001 A1
20020018810 Oshlack et al. Feb 2002 A1
20020031798 Anazawa et al. Mar 2002 A1
20020044968 Van Lengerich Apr 2002 A1
20020081331 Tanner et al. Jun 2002 A1
20020128240 Mazess Sep 2002 A1
20020155154 Wong et al. Oct 2002 A1
20020183288 Mazess et al. Dec 2002 A1
20030059471 Compton et al. Mar 2003 A1
20030083360 Crotts et al. May 2003 A1
20030129194 Mazess et al. Jul 2003 A1
20030138482 Fonkwe et al. Jul 2003 A1
20030152629 Shefer et al. Aug 2003 A1
20030157560 Cantor Aug 2003 A1
20030191093 Chen et al. Oct 2003 A1
20030195171 Daifotis et al. Oct 2003 A1
20040043971 Mazess et al. Mar 2004 A1
20040092534 Yam et al. May 2004 A1
20040101554 Kirschner et al. May 2004 A1
20040132695 Posner et al. Jul 2004 A1
20040197407 Subramanian et al. Oct 2004 A1
20040224930 Posner et al. Nov 2004 A1
20040258749 Guldner et al. Dec 2004 A1
20050014211 Armbruster et al. Jan 2005 A1
20050019374 Modliszewski et al. Jan 2005 A1
20050037064 Basquin et al. Feb 2005 A1
20050069579 Kamaguchi et al. Mar 2005 A1
20050101576 Whitehouse et al. May 2005 A1
20050106233 Andersen et al. May 2005 A1
20050124591 Tian et al. Jun 2005 A1
20050143358 DeLuca et al. Jun 2005 A1
20050147669 Lawrence et al. Jul 2005 A1
20050148557 Tian et al. Jul 2005 A1
20050148558 Knutson et al. Jul 2005 A1
20050186268 Hoshi et al. Aug 2005 A1
20050208055 Chuang et al. Sep 2005 A1
20050287213 Wong et al. Dec 2005 A1
20060009425 Delgado-Herrera et al. Jan 2006 A1
20060019933 Boardman et al. Jan 2006 A1
20060029660 Fonkwe et al. Feb 2006 A1
20060034937 Patel Feb 2006 A1
20060057201 Bonney et al. Mar 2006 A1
20060193877 Tengler et al. Aug 2006 A1
20060210610 Davidson et al. Sep 2006 A1
20060223119 Cantor Oct 2006 A1
20060228808 Clarke et al. Oct 2006 A1
20060257481 Gurney et al. Nov 2006 A1
20070026067 Yam et al. Feb 2007 A1
20070027120 Whitehouse et al. Feb 2007 A1
20070032461 Adorini et al. Feb 2007 A1
20070122477 Bishop et al. May 2007 A1
20070155664 Ranklove et al. Jul 2007 A1
20070190146 Roger et al. Aug 2007 A1
20070207488 Trump et al. Sep 2007 A1
20080109983 Davis May 2008 A1
20080134937 Yang Jun 2008 A1
20080199534 Goldberg et al. Aug 2008 A1
20080317764 Huber et al. Dec 2008 A1
20090004284 Cheng et al. Jan 2009 A1
20090069389 Choi et al. Mar 2009 A1
20090137536 Mazess et al. May 2009 A1
20090155355 Heuer et al. Jun 2009 A1
20090176748 Tabash et al. Jul 2009 A1
20090209501 Bishop et al. Aug 2009 A1
20090262685 Schuringa et al. Oct 2009 A1
20090311316 Bishop et al. Dec 2009 A1
20100120728 Petkovich et al. May 2010 A1
20100144684 Bishop Jun 2010 A1
20100204189 Petkovich et al. Aug 2010 A1
20100227889 Gerspacher et al. Sep 2010 A1
20100291191 Shoichet et al. Nov 2010 A1
20100291197 Schwab Nov 2010 A1
20110039809 Buck et al. Feb 2011 A1
20110039810 Buck et al. Feb 2011 A1
20110039811 Buck et al. Feb 2011 A1
20110105444 Deluca May 2011 A1
20110118218 Buck et al. May 2011 A1
20110130370 Briault et al. Jun 2011 A1
20110171298 Cao Jul 2011 A1
20110182986 Speirs et al. Jul 2011 A1
20110256230 Haeusler et al. Oct 2011 A1
20110300210 Swanson et al. Dec 2011 A1
20110318321 Selva et al. Dec 2011 A1
20110319503 Muller et al. Dec 2011 A1
20120015916 Tabash Jan 2012 A1
20120135103 Walsh et al. May 2012 A1
20130085121 Wang et al. Apr 2013 A1
20130137663 Messner et al. May 2013 A1
20130178451 Bishop et al. Jul 2013 A1
20130189522 Fujii et al. Jul 2013 A1
20130216618 Muller et al. Aug 2013 A1
20130302309 Yang Nov 2013 A1
20140088202 Cade et al. Mar 2014 A1
20140248400 Phonchareon et al. Sep 2014 A1
20140274977 Bishop et al. Sep 2014 A1
20140349979 White et al. Nov 2014 A1
20140357603 Bishop et al. Dec 2014 A1
20150079165 Bishop et al. Mar 2015 A1
20150119472 Shuai et al. Apr 2015 A1
20150119473 Shuai et al. Apr 2015 A1
20170119677 Bishop et al. May 2017 A1
20180021354 Petkovich et al. Jan 2018 A1
Foreign Referenced Citations (134)
Number Date Country
2241205 Jul 1997 CA
101668517 Mar 2010 CN
20321698 Dec 2008 DE
0 227 836 Jul 1987 EP
0413828 Feb 1991 EP
0 508 756 Oct 1992 EP
0 387 808 May 1993 EP
0629405 Dec 1994 EP
1080055 Mar 2001 EP
1208843 May 2002 EP
1 165 061 Oct 2005 EP
1980255 Oct 2008 EP
2 148 661 Dec 2012 EP
2591354 May 2013 EP
2037936 Jun 2014 EP
55-139320 Oct 1980 JP
57-188520 Nov 1982 JP
58-032823 Feb 1983 JP
58-206524 Dec 1983 JP
64-031722 Feb 1989 JP
02-229115 Sep 1990 JP
04-198129 Jul 1992 JP
04-208225 Jul 1992 JP
H04288016 Oct 1992 JP
07-242550 Sep 1995 JP
08-092098 Apr 1996 JP
10-158171 Jun 1998 JP
11-158074 Jun 1999 JP
2001-512418 Aug 2001 JP
2002-302447 Oct 2002 JP
2004-175750 Jun 2004 JP
2004-531548 Oct 2004 JP
2005-505589 Feb 2005 JP
2005-513419 May 2005 JP
2005-528383 Sep 2005 JP
2005-531532 Oct 2005 JP
2005-535682 Nov 2005 JP
2005-538189 Dec 2005 JP
2006-517593 Jul 2006 JP
2006-523221 Oct 2006 JP
2007-525472 Sep 2007 JP
2010-506520 Feb 2010 JP
2010-525079 Jul 2010 JP
2011-512343 Apr 2011 JP
2012-515738 Jul 2012 JP
10-2012-0005228 Jan 2012 KR
WO-91012807 Sep 1991 WO
WO-91016899 Nov 1991 WO
9209271 Jun 1992 WO
WO-94000128 Jan 1994 WO
WO-96000074 Jan 1996 WO
WO-96001621 Jan 1996 WO
WO-96031215 Oct 1996 WO
WO-97011053 Mar 1997 WO
WO-98018610 May 1998 WO
WO-98029105 Jul 1998 WO
WO-99011272 Mar 1999 WO
9949027 Sep 1999 WO
9961398 Dec 1999 WO
WO-00021504 Apr 2000 WO
WO-00035419 Jun 2000 WO
0060109 Oct 2000 WO
WO-00061123 Oct 2000 WO
WO-01037808 May 2001 WO
0172286 Oct 2001 WO
0292056 Nov 2002 WO
0309572 Jan 2003 WO
0330869 Apr 2003 WO
WO-03039521 May 2003 WO
WO-03039572 May 2003 WO
0345381 Jun 2003 WO
WO-03047595 Jun 2003 WO
0386267 Oct 2003 WO
0386415 Oct 2003 WO
WO-03088976 Oct 2003 WO
0393459 Nov 2003 WO
2003106411 Dec 2003 WO
2004010981 Feb 2004 WO
WO-2004028515 Apr 2004 WO
2004054968 Jul 2004 WO
WO-2004058235 Jul 2004 WO
2004071497 Aug 2004 WO
WO-2004080467 Sep 2004 WO
2004098617 Nov 2004 WO
WO-2004101554 Nov 2004 WO
2004110381 Dec 2004 WO
WO-2004110391 Dec 2004 WO
2005000268 Jan 2005 WO
2005003358 Jan 2005 WO
WO-2005011652 Feb 2005 WO
WO-2005123120 Dec 2005 WO
WO-2006052452 May 2006 WO
WO-2006059180 Jun 2006 WO
WO-2006113505 Oct 2006 WO
WO-2007039193 Apr 2007 WO
WO-2007039569 Apr 2007 WO
WO-2007047327 Apr 2007 WO
WO-2007050724 May 2007 WO
WO-2007050975 May 2007 WO
WO-2007053608 May 2007 WO
WO-2007068287 Jun 2007 WO
2007092221 Aug 2007 WO
WO-2007092755 Aug 2007 WO
WO-2007146004 Dec 2007 WO
WO-2008008608 Jan 2008 WO
WO-2008043449 Apr 2008 WO
WO-2008097646 Aug 2008 WO
2008116113 Sep 2008 WO
2008116133 Sep 2008 WO
2008134518 Nov 2008 WO
WO-2008134512 Nov 2008 WO
WO-2008134523 Nov 2008 WO
WO-2009047644 Apr 2009 WO
2009101135 Aug 2009 WO
WO-2009101132 Aug 2009 WO
WO-2009101137 Aug 2009 WO
2009124210 Oct 2009 WO
WO-2010011906 Jan 2010 WO
WO-2010034342 Apr 2010 WO
WO-2011031621 Mar 2011 WO
2011063952 Jun 2011 WO
WO-2011095388 Aug 2011 WO
2011123476 Oct 2011 WO
2012006475 Jan 2012 WO
WO-2012018329 Feb 2012 WO
WO-2012076429 Jun 2012 WO
WO-2012091569 Jul 2012 WO
WO-201 2117236 Sep 2012 WO
2012145491 Oct 2012 WO
2014029953 Feb 2014 WO
2014143941 Sep 2014 WO
2014193255 Dec 2014 WO
WO-2014202754 Dec 2014 WO
2016020508 Feb 2016 WO
Non-Patent Literature Citations (367)
Entry
Biochemical Effects of Calcium and Vitamin D Supplementation in elderly, institutionalized, vitamin D-deficient Patients, Rev. Rhum. [Engl. Ed. 63 (2), 135-140), Feb. 1996.
Halloran J. clin. Endocrinal. Metabolism, Dec. 1984 59(6) pp. 1063-1069).
Zerwekh et al (Extra-renal production of 24,25-dihydroxyvitamin D in chronic renal failure during 25 hydroxyvitamin D3 therapy, Kidney International, vol. 23, (1983), pp. 401-406).
Zerwekh et al. (Kidney International, vol. 23, (1983), pp. 401-406).
International Preliminary Reporton Patentability for corresponding international application No. PCT/US2011/030404, dated Oct. 2, 2012.
International Search Report and Written Opinion for corresponding international application No. PCT/US11/30404, dated May 25, 2011.
Kidney Disease Improving Global Outcomes (KDIGO) Clinical Practice Guidelines for Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease—Mineral and Bone Disorder (CKD-MBD), Kidney International Supplement, 113:S1-130 (2009).
Kidney Disease Outcomes Quality Inititiative (K/DOQI) Clinical Practice Guidelines for Bone Metabolism and Disease in Chronic Kidney Disease, Am. J. Kidney Dis., 42:S1-S202 (2003).
Standing Committee on the Scientific Evaluation of Dietary Reference Intakes, Dietary reference intakes: calcium, phosphorous, magnesium, vitamin D, and fluoride, Washington, DC: National Academy Press (1997).
Kidney Disease Improving Global Outcomes (KDIGO) Clinical Practice Guidelines for Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease—Mineral and Bone Disorder (CKD-MBD), Kidney International Supplement, 113:S1-130 (2009) at S70-S89 (Chapter 4.2: Treatment of Abnormal PTH Levels in CKD-MBD).
Kidney Disease Outcomes Quality Inititiative (K/DOQI) Clinical Practice Guidelines for Bone Metabolism and Disease in Chronic Kidney Disease, Am. J. Kidney Dis., 42:S1-S202 (2003) at S52-S57 (Guideline 1. Evaluation of Calcium and Phosphorus Metabolism).
Standing Committee on the Scientific Evaluation of Dietary Reference Intakes, Dietary reference intakes: calcium, phosphorous, magnesium, vitamin D, and fluoride, Washington, DC: National Academy Press (1997) at 250-287 (Chapter 7: Vitamin D).
Baez et al., Hipocalcemia severa posdenosumab, Nefrologfa (Madrid), 33(4): 614-615 (2013).
Baker et al., Plasma 25-hydroxy vitamin D concentrations in patients with fractures of the femoral neck, British Medical Journal, 1(6163):589 (1979).
Gradishar et al., Minimizing cancer's impacton bone with denosumab: current and future perspectives, Community oncology, 10(8):235-243 (2013).
Melanie S Joy Pharmd FCCP et al.: “Outcomes of Secondary Hyperparathyroidism in Chronic Kidney Disease and the Direct Costs of Treatment”, Journal of Managed Care Pharmacy, Academy of, Managed Care Pharmacy, Alexandria, VA, vol. 13, No. 5, Jan. 1, 2007 (Jan. 1, 2007), p. 397^11.
Sirvent et al., Extreme hypocalcaemia and hyperparathyroidism following denosumab. Is this drug safe in chronic kidney disease?, Nefrologfa (Madrid), 34(4): 542-544 (2014).
Jones, “Why dialysis patients need combination therapy with both cholecalciferol and a calcitriol analogs,” Seminars in Di alysis, pp. 1-5 (2010).
Kaufmann et al. J. Clin. Endocrinol. Metab., 2014, vol. 99, pp. 2567-2574 (Year: 2014).
Kaufmann et al., Clinical utility of simultaneous quantitation of 25-hydroxyvitamin D and 24,25-dihydroxyvitamin D by LC-MS/MS involving derivatization with DMEQ-TAD, J. Clin. Endocrinol. Metab., 99(7):2567-74 (Jul. 2014).
Kazama et al., Role of circulating fibroblast growth factor 23 in the development of secondary hyperparathyroidism, Ther. Apher. Dial., 9:328-30 (2005).
KDOQI Clinical practice guidelines 2004. National Kidney Foundation).
Kidney Disease Improving Global Outcomes (KDIGO) 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease—Mineral and Bone Disorder (CKD-MBD). Kidney Int Suppl. 2017;7(1):1-59.
Kovesdy et al., Association of activated vitamin D treatment and mortality in chronic kidney disease, Arch. Intern. Med., 168(4):397-403 (Feb. 2008).
Krishnan et al., The role of vitamin D in cancer prevention and treatment, Rheum. Dis. Clin. North Am., 38(1):161-78 (2012).
KURO-O, Klotho in chronic kidney disease—what's new?, Nephrol. Dial. Transplant., 4 pp. (2009).
Lo et al., Vitamin D absorption in healthy subjects and in patients with intestinal malabsorption syndromes, Am. J. Clin. Nutr., 42(4):644-9 (1985).
Luo et al., 24-Hydroxylase in cancer: impact on vitamin D-based anticancer therapeutics, J. Steroid Biochem. Mol. Biol., 136:252-7 (2013).
M. Larrosa et al. (Abstract FR10365, Long term treatment of Hypovitaminisis, clacidiol or cholecalcidiol).
Martin-Baez et al., Severe hypocalcaemia post-denosumab, Nefrologia, 33(4):614-5 (2013).
Mimori et al., Clinical significance of the overexpression of the candidate oncogene CYP24 in esophageal cancer, Ann. Oncol., 15(2):236-41 (2004).
Motelion et al., Parathyroid hormone-related protein, parathyroid hormone, and vitamin D in hypercalcemia of malignancy, Clin. Chim. Acta, 290(2):189-97 (2000).
NASMHPD Medical Director's Technical Reporton Psychiatric Polypharmacy, Sep. 2001.
National Kidney Foundation Guidelines, NKF, Am. J. Kidney Dis., 42(4,Suppl 3):S1-S202 (2003).
NewsWire (https://www.newswire.ca/news-releases/cytochroma-announces-data-presentations-at-american-society-of-hephrologys43rd-annual-meeting-and-scientific-exposition-546289852.html, published Nov. 18, 2010) (Year: 2010).
NewsWire (https://www.newswire.ca/news-releases/cytochroma-announces-data-presentations-at-american-society-of-nephrologys43rd-annual-meeting-and-scientific-exposition-546289852.html, published Nov. 18, 2010) (Year: 2010).
Non-Final Office Action received for U.S. Appl. No. 12/597,230, dated Dec. 13, 2019, 15 pages.
Notice of Allowance, U.S. Appl. No. 15/918,620, dated Nov. 29, 2019, 9 pages.
Olmos et al., Effects of 25-hydroxyvitamin D3 therapy on bone turnover markers and PTH levels in postmenopausal osteoporotic women treated with alendronate, J. Clin Endocrinol. Metab., 97(12):4491-7 (2012).
OPKO Health Inc., Safety/Efficacy Study of CTAP101 in Chronic Kidney Disease Subjects With Secondary Hyperparathyroidism (SHPT), <https://clinicaltrials.gov/ct2/show/NCT01219855> Oct. 13, 2010.
Package insert for Hectorol (doxercalciferol capsules), Genzyme (2011).
Package insert for Zemplar (paricalcitol) Capsules, Abbott (2011).
Pak et al., “Treatment of Vitamin D-Resistant Rickets With 25-Hydroxycholecalciferol,” Arch Intern Med, 129:894-899 (1972).
Parise et al., CYP24, the enzyme that catabolizes the antiproliferative agent vitamin D, is increased in lung cancer, Int. J. Cancer, 119(8):1819-28 (2006).
Petkovich et al., “CYP24A1 and Kidney Disease,” Current Opin. in Nephrology and Hypertension, 20:337-344 (2011).
Posner et al., “Vitamin D Analogues Targeting CYP24 in Chronic Kidney Disease,” J. Steroid Biochem and Mol. Biol., 121:13-19(2010).
Querfeld et al., Vitamin D deficiency and toxicity in chronic kidney disease: in search of the therapeutic window, Pediatr. Nephrol., 25(12):2413-30 (Dec. 2010).
Rabbani, Molecular mechanism of action of parathyroid hormone related peptide in hypercalcemia of malignancy: therapeutic strategies (review), Int. J. Oncol., 16(1):197-206 (2000).
Ravani et al., Vitamin D levels and patient outcome in chronic kidney disease, Kidney Int., 75(1):88-95 (Jan. 2009).
Richard et al., PTHrP gene expression in cancer: do all paths lead to Ets?, Crit. Rev. Eukaryot. Gene Expr., 15(2):115-32 (2005).
Rocaltrol (Registered) Complete Product Information, Roche, Jul. 27, 2004.
Saseen et al., “Dual calcium-channel blocker therapy in the treatment of hypertension,” Ann Pharmacother., 30(7-8):802-10 (1996).
Sato et al., Increased 1,25-(OH)2D2 concentration in a patient with malignancy-associated hypercalcemia receiving intravenous hyperalimentation inadvertently supplemented with vitamin D2, Intern. Med., 32(11):886-90 (1993).
Schwariz et al., Extended-release calcifediol (ERC) effectively increased serum 25-hydroxyvitamin D levels in breast and prostate cancer patients without significant impact on serum calcium or phosphorus, Opko Renal (2018).
Segersten et al.: Potentiating effects of nonactive/active vitamin D analogues and ketoconazole in parathyroid cells, Clinical Endocrinology., vol. 66, No. 3, Mar. 1, 2007 (Mar. 1, 2007), pp. 399-404.
Sensipar (cinacalcet) prescriptioninformation, revised Aug. 2011.
Sensipar package insert (Year: 2004).
Sicinski et al., “Synthesis of 1 alpha, 25-Dihydroxyvitamin D2, Its 24 Epimer and Related Isomers, and Their Binding Affinity for the 1, 25-Dihydroxyvitamin D3 Receptor,” Bioorganic Chemistry, 13: 158-169 (1985).
Skugor M. et al.: Ëvolution and current state of assays for parathyriod hormone, Biochemia Medica, vol. 20, No. 2, 2010, pp. 221-228.
Soyfoo et al., Non-malignant causes of hypercalcemia in cancer patients: a frequent and neglected occurrence, Support Care Cancer, 21(5):1415-9 (2013).
Sprague et al., Use of Extended-Release Calcifediol to Treat Secondary Hyperparathyroidism in Stages 3 and 4 Chronic Kidney Disease, Am. J. Nephrol., 44(4):316-25 (2016).
Stavroulopoulos et al., Relationship between vitamin D status, parathyroid hormone levels and bone mineral density in patients with chronic kidney disease stages 3 and 4, Nephrology (Carlton), 13(1):63-7 (Feb. 2008).
Tamez et al., Vitamin D reduces left atrial volume in patients with left ventricular hypertrophy and chronic kidney disease, Am. Heart J., 164(6):902-9.e2 (Dec. 2012).
Tebben et al., Elevated fibroblast growth factor 23 in women with malignant ovarian tumors, Mayo Clin. Proc., 80:745-51 (2005).
Terrie, Monitoring Combination Drug Therapy, Pharmacy Times, published Jan. 18, 2010., Jan. 19, 2010.
Tomida et al., Serum 25-hydroxyvitamin D as an independent determinant of 1-84 PTH and bone mineral density in non-diabetic predialysis CKD patients, Bone, 44(4):678 83 (Apr. 2009).
Tsuji, et al. “A New and Convenient Synthesis of 1a,25-Dihydroxyvitamin D2 and It 24R-Epimer,” Bull. Chem. Soc. Jpn., 62:10 pp. 3132-3137 (1989).
“Hidroferol® (calcifediol): Casos de Hipercalcemia e Hipervitaminosis D,” Butlletí de Farmacovigilància de Catalunya, 9(5):17-20 (2011).
“Modern Pharmaceutics” 4th ed., Marcel Dekker, Inc., New York, NY, p. 16-21 (2002).
Al-Aly, Z., “Changes in Serum 25-Hydroxyvitamin D and Plasma Intact PTH Levels Following Treatment with Ergocalciferol in Patients With CKD,” Am. J. Kid. Dis., 50(1):59-68 (2007).
Alvarez et al., “Vitamin D Supplementation in Pre-Dialysis Chronic Kidney Disease,” Dermato-Endocrinology, 4(2):118-127 (2012).
Andress, “Vitamin D in chronic kidney disease: A systematic role for selective vitamin D receptor activation,” Kidney Int., 69:33-43 (2006).
Arekat et al., “Dramatic Improvement of BMD Following Vitamin D Therapy in a Bone Marrow Transplant Recipient,” J. Clin. Densitometry, 5:267-271 (2002).
Armas et al., “Vitamin D2 is Much Less Effective than Vitamin D3 in Humans,” J. Clin. Endocrinol. Metab., 89:5387-5391 (2004).
Bagnis et al., “Biochemical and Hormonal Short-Term Effects of 25-hydroxyvitamin D3 in Patients on Continuous Peritoneal Dialysis,” Ital. J. Mineral Electrolyte Metab., 12:73-76 (1998).
Bailie et al. “Comparative Review of the Pharmacokinetics of Vitamin D Analogues,” Seminars in Dialysis, 15(5):352-357 (2000).
Baird et al., “Steroid Dynamics Under Steady-State Conditions,” Recent Prog. Harm. Res., 25:611-664 (1969).
Barger-Lux M.J. et al., “Vitamin D and Its Major Metabolites: Serum Levels After Graded Oral Dosing In Healthy Men” Osteoporosis International, United Kingdom, 8(3):222-230 (1998).
Barreto et al., “25-Hydroxyvitamin D3, the Prohormone of 1,25-Dihydroxyvitamin D3, Inhibits the Proliferation of Primary Prostatic Epithelial Cells,” Cancer Epidemiol, Biomarkers & Prevention, 9:265-270 (2000).
Beckman, et al., “Up-Regulation of the Intestinal 1, 25-Dihydroxyvitamin D Receptor During Hypervitaminosis D: A Comparison Between Vitamin D2 and Vitamin D31,” Biochemical and Biophysical Research Communications, vol. 169, No. 3, pp. 910-915 (Jun. 29, 1990).
Beer et al., “Pharmacokinetics and Tolerability of a Single Dose of DN-101, a New Formulation of Calcitriol, in Patients with Cancer,” Clin. Cancer Res., 11:7794-7799 (2005).
Bell et al., “Evidence that 1,25-Dihydroxyvitamin D3 Inhibits the Hepatic Production of 25-Hydroxyvitamin D in Man,” J. Clin. Invest., 74:1540-1544 (1984).
Belostotsky et al., “A single high dose of ergocalciferol can be used to boost 25-hydroxyvitamin D levels in children with kidney disease,” Pediatr Nephrol, 24:625-626 (2009).
Bianchi et al., “No Difference in Intestinal Strontium Absorption After an Oral or an Intravenous 1,25(OH)2D3 Bolus in Normal Subjects,” J. Bone Miner. Res., 14:1789-1795 (1999).
Binkley et al., “Laboratory Reporting of 25-Hydroxyvitamin D Results: Potential for Clinical Misinterpretation,” Clinical Chemistry, 52(11);2124-2125 (2006).
Blair et al., “Prevalence of vitamin D [25(OH)D] deficiency and effects of supplementation with ergocalciferol (vitamin D2) in stage 5 chronic kidney disease patients.” J.Ren Nutr., 18: 375-382 (2008).
Bordier et al., “Evolution of renal osteodystrophy: Correlation of bone histomorphometry and serum mineral and immunoreactive parathyroid hormone values before and after treatment with calcium carbonate or 25-hydroxycholecalciferol,” Kidney Int Suppl, 2:S102-S112 (1975).
Boudville et al., “Renal Function and 25-Hydroxyvitamin D Concentrations Predict Parathyroid Hormone Levels in Renal Transplant Patients,” Nephrol Dial Transplant, 21:2621-2624 (2006).
Bouillon et al., “Influence of dialysate calcium concentration and vitamin D on serum parathyroid hormone during repetitive dialysis,” Kidney Int., 7:422-432 (1975).
Brossard et al. “Influence of Glomerular Filtration Rate on Non-(1-84) Parathyroid Hormone (PTH) Detected by Intact PTH Assays,” Clinical Chemistry, 46(5):697-703 (2000).
Brown et al., “The Vitamin D Prodrugs 1α(OH)D2, 1α(OH)D3 and BCI-210 Suppress PTH Secretion by Bovine Parathyroid Cells,” Nephrol Dial Transplant, 21:644-650 (2006).
Brown et al., “Vitamin D Analogues for Secondary Hyperparathyroidism,” Nephrol Dial Transplant, 17[Suppl. 10]:10-19 (2002).
Buccianti et al., “Effects of Calcifediol Treatment on the Progression of Renal Osteodystrophy during Continuous Ambulatory Peritoneal Dialysis,” Nephron, 56:353-356 (1990).
Budavari (ed.), Merck Index: an Encyclopedia of Chemicals, Drugs, and Biologicals, 11th Edition, Merck & Co., 9927-9930 (1989).
Bulla et al., “Renal bone disorders in children: therapy with vitamin D3 or 1,25-dihydroxycholecalciferol,” Proc.Eur.Dial.Transplant.Assoc., 16: 644-648 (1979).
Chandra et al., “Cholecalciferol (vitamin D3) therapy and vitamin D insufficiency in patients with chronic kidney disease: a randomized controlled pilot study,” Endocr.Pract., 14: 10-17 (2008).
Claris-Appiani et al., “Phosphate-Induced PTH Stimulation and Calcitriol Treatment in Children with Early Chronic Renal Insufficiency,” J. Bone Miner. Met., 12:S91-S97 (1994).
Coburn et al., “Doxercalciferol Safely Suppresses PTH Levels in Patients with Secondary Hyperparathyroidism Associated with Chronic Kidney Disease Stages 3 and 4,” Am. J. Kidney Dis., 43(5):877-890 (2004).
Coburn, “An Update on Vitamin D as Related to Nephrology Practice: 2003,” Kidney International, vol. 64, Supplement 87, pp. S125-S130 (2003).
Coburn, et al., “Use of Active Vitamin D Sterols in Patients with Chronic Kidney Disease, Stages 3 and 5,” Kidney International, vol. 63, Supplement 85, pp. S49-S53 (2003).
Coen et al., “1,25(OH)2D3 and 25-OHD3 in the Treatment of Renal Osteodystrophy: Comparison of Combined Versus 1,25(OH)2D3 Administration Alone,” Miner. Electrolyte Metab., 9:19-27 (1983).
Coen et al., “25-hydroxycholecalciferol in the treatment of renal osteodystrophy in haemodialysed patients,” Int J Artificial Organs, 2(6): 278-281 (1979).
Cohen-Solal et al., “Non-Aluminic Adynamic Bone Disease in Non-Dialyzed Uremic Patients: A New Type of Osteopathy Due to Overtreatment?” Bone, 13:1-5 (1992).
Collet et al. “Modified-Release Peroral Dosage Forms,” Aulton (ed.), Pharmaceutics: The Science of Dosage Forms, Churchill Livingston, London, pp. 289-305 (2002).
Colodro et al., “Effect of 25-Hydroxy-Vitamin D3 on Intestinal Absorption of Calcium in Normal Man and Patients With Renal Failure,” Metabolism, 27(6):745-753 (1978).
Cooke et al., “Vitamin D-Binding Protein (Gc-Globulin): Update 1995,” Endocrine Rev., 4:125-128 (1995).
Coyne et al., “Paricalcitol Capsule for the Treatment of Secondary Hyperparathyroidism in Stages 3 and 4 CKD,” American Journal of Kidney Diseases, 47(2):263-276 (2006).
Daisley-Kydd et al., “Calcitriol in the Management of Secondary Hyperparathyroidism of Renal Failure,” Pharmacotherapy., 16:619-630 (1996).
Davies, M. et al. The Absorption and Metabolism of Vitamin D3 from Parenteral Injection Sites, Proceedings of the Workshop on Vitamin D, 4th, Vitam. D: Basic Res. Its Clin. Appl. (1979), abstract.
DB-Pharma, “Dedrogyl 15 Mg/10ML Calcifediol Oral Drops, Solution,” Marketing Authorization No. 317 863.2 (2000).
DeLuca, “Treatment of renal osteodystrophy with 25-hydroxycholecalciferol,” Arch Intern Med, 126(5):896-899(1970).
Deroisy et al., “Comparison of the Short-Term Effects of Three Oral Calcium-Vitamin D Formulations and Placebo on Calcium Metabolism,” Curr. Ther. Res., 59:370-378 (1998).
DeVille et al., “Effect of Ergocalciferol Supplementation on Serum Parathyroid Hormone and Serum 25-Hydroxyvitamin D in Chronic Kidney Disease,” Nephrology, 11:555-559 (2006).
Dietary Reference Intakes for Calcium, Phosphorus, Magnesium, Vitamin D, and Fluoride, Standing Committee on the Scientific Evaluation of Dietary Reference Intakes, Food and Nutrition Board, Institute of Medicine, National Academy Press, Washington DC, pp. 250-287 (1997).
Dietary Supplement Fact Sheet: Vitamin D, National Institutes of Health, Office of Dietary Supplements (last update: Aug. 5, 2005), retrieved from <URL: http:ods.od.nih.gov/factsheets/vitamind.asp> on Aug. 31, 2007.
Disease and Vitamin D, University of California Riverside, retrieved from Internet, <URL: <http://vitamind.ucr.edu/disease.html>> (last update: May 19, 1999).
Disintegration, chapter 701; Dissolution, chapter 711; Distilling Range, chapter 721; Drug Release, chapter 724; Electrophoresis, chapter 726; pp. 276-292, in: U.S. Pharmacopeia vol. 30.
Dogan et al., “Effect of depot oral cholecalciferol treatment on secondary hyperparathyroidism in stage 3 and stage 4 chronic kidney diseases patients,” Ren Fail., 30: 407-410 (2008).
Drueke et al., Recurrence of hyperparathyroidism from autografted parathyroid fragments in uremic patients in spite of administration of 25(OH)D3 and 1a(OH)D3. In: Vitamin D. Basic Research and its Clinical Application, (Eds. Norman AW, Schaefer K, Herrath Dv, Grigoleit HG, Coburn JW, DeLuca HF, Mawer EB, and Suda T), pp. 791-794. Willem de Gruyter, New York (1979).
Dusso et al., “Metabolic Clearance Rate and Production Rate of Calcitriol in Uremia,” Kidney Int., 35 860-864(1989).
Dusso et al., “Extra-renal production of calcitriol in chronic renal failure,” Kidney Int., 34:368-375 (1988).
Dusso et al., “Extrarenal Production of Calcitrol in Normal and Uremic Humans*,” Journal of Clinical Endocrinology and Metabolism, 72(1):157-164 (1991).
Eastwood et al., “Biochemical and histological effects of 1,25 dihydroxycholecalciferol (1,25-DHCC) in the osteomalacia of chronic renal failure,” J Urol Nephrol (Paris,) 80(12): 984-985 (1974).
Eastwood et al., “The contrasting effects on bone histology of vitamin D and of calcium carbonate in the osteomalacia of chronic renal failure,” Clin Sci Molec Med, 47:23-42 (1974).
Eastwood et al., “The Effect of 25-Hydroxy Vitamin D3 in the Osteomalacia of Chronic Renal Failure,” Clin. Sci. Molec. Med., 52:499-508 (1977).
Fernandez et al., “Guidelines for Dosing of Intravenous Calcitriol in Dialysis Patients with Hyperparathyroidism,” Nephrol. Dial. Transplant., 11:96-101 (1996).
Fournier et al., “1α Hydroxycholecalciferol and 25 Hydroxycholecalciferol in Renal Bone Disease” Proc Eur Dial Transplant Assoc 12:227-236 (1976).
Fournier et al., “Advances in Nephrology from the Necker Hospital” Adv. Nephrol Necker Hosp. 21:237-306 (1992).
Fournier et al., “Comparison of 1α-hydroxycholecalciferol and 25-hydroxycholecalciferol in the treatment of renal osteodystrophy: Greater effect of 25-hydroxycholecalciferol on bone mineralization” Kidney International 15:196-204 (1979).
Fournier et al., “Current Status of the Management of Renal Osteodystrophy” Proceedings of the European Dialysis and Transplant Association 15:547-568 (1978).
Fournier et al., “Importance of Vitamin D Repletion in Uraemia,” Nephrol Dial Transplant, 14(4):819-823 (1999).
Fournier et al., “Low doses of calcitriol or calcium carbonate for the prevention of hyperparathyroidism in predialysis patients?” Nephrol Dial Transplant 11 (7):1493-1495 (1996).
Fournier et al., “Present-Day Concepts in the Treatment of Chronic Renal Failure” Contrib Nephrol. 71:64-80 (1989).
Fournier et al., “Preventing Renal Bone Disease in Moderate Renal Failure with CaCO3 and 25(OH) Vitamin D3,” Kidney Int., 33:S178-S279 (1988).
Fournier et al., “Renal Osteodystrophy in Dialysis Patients: Diagnosis and Treatment,” Artificial Organs, 22:530-557 (1998).
Fournier et al., “Renal Osteodystrophy: Pathophysiology and Treatment” Hormone Res. 20:44-58 (1984).
Fournier et al., “The Approach to the Treatment of Secondary Hyperparathyroidism in Early Renal Failure” Am. J. Nephrol 8:170-172 (1988).
Fournier et al., “Traitement vitaminique D et ostéodystrophies rénales: indications et modalitiés” Nephrologie 16(2):165-190 (1995) [journal in French].
Fournier, “Vitamin D: Biochemical, Chemical, and Clinical Aspects Related to Calcium Metabolism,” Vitamin D: Proceedings of the Third Workshop on Vitamin D, Asilomar, Pacific Grove, CA, USA 667-669 (1977).
Friedman et al. “The Role of Vitamin D in Mild to Moderate Chronic Kidney Disease,” Trends in Endocrinology & Metab,. 13(5):189-194 (2002).
Fritsche et al., “Regulation of 25-Hydroxyvitamin D3-1α-Hydroxylase and Production of 1 α,25-Dihydroxyvitamin D3 by Human Dendritic Cells,” Blood, 102(9):3314-3316 (2003).
Frohling et al., “Serum 25-hydroxyvitamin D in patients with chronic renal failure on long-term treatment with high doses of vitamin D2.” Nephron 26: 116-120 (1980).
Frost et al., “Histomorphometric Changes in Trabecular Bone of Renal Failure Patients Treated with Calcifediol,” Metab. Bone Dis. & Rel. Res., 2:285-295 (1981).
Gallagher et al., “Comparison of the Histological Effect and Metabolism of 25-(OH)D and 1,25-(OH)2D in Rat Bone,” p. 399-401, In: Norman, Vitamin D: Basic Research and its Clinical Application: Proceedings of the Fourth Workshop on Vitamin D, Berlin, West Germany, Feb. 1979.
Ghazali et al., “Is low plasma 25-(OH) vitamin D a major risk factor for hyperparathyroidism and Looser's zones independent of calcitriol?” Kidney International 55:2169-2177 (1999).
Gibson, ed., Product optimisation. Pharmaceutical Preformulation and Formulation: A Practical Guide from Candidate Drug Selection to Commercial Dosage Form, 295-8 (2004).
Granja et al., “Studies on the Opening of Dioxanone and Acetal Templates and Application to the Synthesis of 1α,25-Dihydroxyvitamin D21,” J. Org. Chem., 58:124-131 (1993).
Gómez-Alonso et al., “Vitamin D Status and Secondary Hyperparathyroidism: The Importance of 25-Hydroxyvitamin D Cut-Off Levels,” Kidney International, 63(Supp. 85):S44-S48 (2003).
Haddad et al., “Human Serum Binding Capacity and Affinity for 25-Hydroxyergocalciferol and 25-Hydroxycholecalciferol,” J. Clin. Endocrinol. Metab., 43:86-91 (1976).
Haddad et al., “Natural and Synthetic Sources of Circulating 25-Hydroxyvitamin D in Man,” Nature, 244:515-517 (1973).
Haddad, “Plasma Vitamin D-binding Protein (Gc-Globulin): Multiple Tasks,” J. Steroid Biochem. Molec. Biol., 53:579-582 (1995).
Haddad, “Seasonal Diminution of Vitamin D Stores in the United States: Can Darker Winters Lead to Lighter Bones?” Trends Endocrinol. Metab., 7:209-212 (1996).
Haddad, “Traffic, Binding and Cellular Access of Vitamin D Sterols,” Bone and Mineral Res., Elsevier, 5:281-308 (1987).
Haddad, “Vitamin D—Solar Rays, The Milky Way, or Both?” NEJM, 326:1213-1215 (1992).
Haldimann et al., “Effect of an Oral Dose of 25-Hydroxyvitamin D3 on Its Blood Levels in Patients with the Nephrotic Syndrome,” J Clin Endocrinology and Metabolism, 50(3): 470-474 (1980).
Halloran et al., “Plasma Vitamin D Metabolite Concentrations in Chronic Renal Failure: Effect of Oral Administration of 25-Hydroxyvitamin D3,” J. Clin. Endocrin. & Metab., 59:1063-1069 (1984).
Hamida et al., “Hyperparathyroïdie secondaire álinsuffisance rénale” Annales d'Endocrin-ologie 55:147-158 (1994) [reference in French].
Hannula et al., “Constant, But Not Pulsed Calcitriol Suppresses Hemodialysis Patients' Antigen-Induced Lymphocyte Proliferation,” Nephron, 86:139-144 (2000).
Hari et al., “Vitamin D insufficiency and effect of cholecalciferol in children with chronic kidney disease,” Pediatr.Nephrol,. 25: 2483-2488 (2010).
Hay et al., “Vitamin D2 in Vertebrate Evolution,” Comp. Biochem. Physiol. B, 56:375-380 (1977).
Hodson et al., “Treatment of childhood renal osteodystrophy with calcitriol or ergocalciferol,” Clin Nephrology, 24(4): 192-200 (1985).
Holick, “Vitamin D Deficiency in CKD: Why Should We Care?” Am. J. Kidney Dis., 45:1119-1121 (2005).
Holick, “Vitamin D Status: Measurement, Interpretation and Clinical Application,” Ann Epidemiol, 19(2):73-78 (2009).
Hollis, “Circulating 25-Hydroxyvitamin D Levels Indicative of Vitamin D Sufficiency: Implications for Establishing a New Effective Dietary Intake Recommendation for Vitamin D,” J. Nutr. 135: 317-322 (2005).
Horst et al., “A Sensitive Competitive Protein Binding Assay for Vitamin D in Plasma,” Steroids, 37:581-592 (1981).
Horst et al., “Discrimination in the Metabolism of Orally Dosed Ergocalciferol and Cholecalciferol by the Pig, Rat and Chick,” Biochem. J., 204:185-189 (1982).
Horst et al., “Rat cytochrome P450C24 (CYP24) does not metabolize 1,25-dihydroxyvitamin D2 to calcitroic acid,” J. Cell Biochem., 88:282-285 (2003).
Hottelart et al., “Ostéodystrophie rénale (2): son traitement chez l'insuffisant rénal avant la dialyse” Nephrologie 21(6):275-282 (2000) [reference in French].
Houghton et al., “The Case Against Ergocalciferol (Vitamin D2) as a Vitamin Supplement,” Am. J. Clin. Nutr., 84:694-697 (2006).
Hunt, et al., “A Comparison of the Toxicity of Ergocalciferol and Cholecalciferol in Rhesus Monkeys (Macaca mulatta),” J. Nutrition, 102:975-986 (1972).
Hussar, “New Drugs of 1999,” J. Am. Pharmacist. Assoc. 40(2):181-229 (2000).
International Search Report for Application No. PCT/US2007/061521, dated Jul. 17, 2007.
International Search Report of PCT/US2008/061579 dated Aug. 21, 2008 (5 pages).
Ishimura et al., “Serum Levels of 1,25-Dihydroxyvitamin D, 24,25-Dihydroxyvitamin D, and 25-Hydroxyvitamin D in Nondialyzed Patients with Chronic Renal Failure,” Kidney Int., 55:1019-1027 (1999).
Japanese Office Action for Application No. 2008-553520, dated Jul. 24, 2013.
Jara et al., “Effect of Calcitriol Treatment and Withdrawal on Hyperparathyroidism in Haemodialysis Patients with Hypocalcaemia,” Nephrol. Dial. Transplant., 16:1009-1016 (2001).
Jean et al., “Daily Oral 25-Hydroxycholecalciferol Supplementation for Vitamin D Deficiency in Haemodialysis Patients: Effects on Mineral Metabolism and Bone Markers,” Nephrol. Dial. Transplant, 23:3670-3676 (2008).
Jean et al., “Evidence for Persistent Vitamin D 1-Alpha-Hydroxylation in Hemodialysis Patients: Evolution of Serum 1,25-Dihydroxycholecalciferol after 6 Months of 25-Hydroxycholecalciferol Treatment” Nephron. Clin. Pract. 110:c58-c65 (2008).
Jean et al., “Monthly cholecalciferol administration in heamodialysis patients: a simple and efficient strategy for vitamin D supplementation” Nephrol. Dial. Transplant 24(12):3799-3805 (2009).
Jones, “Pharmacokinetics of vitamin D toxicity,” Am. J. Clin. Nutr. 88(suppl): 582S-6S (2008).
Jones., “Expanding the Role for Vitamin D in Chronic Kidney Disease: Importance of Blood 25-OH-D Levels and Extra-Renal 1α-Hydroxyase in the Classical and Nonclassical Actions of 1α, 25-Dihydroxyvitamin D3,” Seminars in Dialysis, 20(4):316-324 (2007).
Kajihara et al., “Novel Method to Control Release of Lipophilic Drugs with High Potency from Silicone,” Chem. Pharm. Bull., 51:11-14 (2003).
Kalantar-Zadeh et al., “Clinical Outcomes with Active versus Nutritional Vitamin D Compounds in Chronic Kidney Disease” Clin J Am Soc Nephrol. 4(9):1529-1539 (2009).
Kanis et al., “Rate of Reversal of Hypercalcaemia and Hypercalciuria Induced by Vitamin D and Its 1-alpha-Hydroxylated Derivatives,” BMJ, 1:78-81 (1977).
Khachane et al., “Novel Sustained Release Drug Delivery System: Review,” IJPRD, 3(12):1-14 (2012).
Kim, Advanced Pharmaceutics: Physicochemical Principles, pp. 362-392, Boca Raton, Fla: CRC Press (2004).
Kinoshita et al., “1,25-Dihydroxyvitamin D Suppresses Circulating Levels of Parathyroid Hormone in a Patient with Primary Hyperparathyroidism and Coexistent Sarcoidosis,” J. Clin. Endo. & Metabol., 90(12):6727-6731 (2005).
Kleinman et al., “Effects of Calcifediol on Calcified Tissue in Uremia,” Arch Intern Med, 138: 864-865 (1978).
Kobayashi et al., “2β-(3-Hydroxyproxy)-α, 25-Dihydroxyvitamin D3 (ED-71), Preventive and Therapeutic Effects on Bone Mineral Loss in Ovariectomized Rats,” Bioorganic & Medicinal Chemistry Letters, 3(9):1815-1819 (1993).
Kobayashi et al., “Variation of 25-Hydroxyvitamin D3 and 25-Hydroxyvitamin D2 Levels in Human Plasma Obtained from 758 Japanese Healthy Subjects,” J. Nutr. Sci. Vitaminol (Tokyo), 29(3):271-281 (1983). Abstract Only.
Kooienga et al., “The effect of combined calcium and vitamin D3 supplementation on serum intact parathyroid hormone in moderate CKD,” Am.J.Kidney Dis,. 53: 408-416 (2009).
Koshikawa, et al., “Clinical Effect of Intravenous Calcitriol Administration on Secondary Hyperparathyroidism,” Nephron; 90:413-423 (2002).
LaClair et al., “Prevalence of Calcidiol Deficiency in CKD: A Cross-Sectional Study Across Latitudes in the United States,” Am. J. Kidney Dis., 45:1026-1033 (2005).
Lafage et al., “Ketodiet, Physiological Calcium Intake and Native Vitamin D Improve Renal Osteodystrophy,” Kidney Int., 42:1217-1225 (1992).
Lambert et al., “Evidence for Extranrenal Production of 1-alpha,25-Dihydroxyvitamin D in Man,” J. Clin. Invest., 69:722-725 (1982).
Lambrey et al., “24, 25 Dihydroxycalciferol: Assay in Non-Anephric Patients on Chronic Haemodialysis and Assessment of it's Possible Pathophysiological Role in Renal Osteodystrophy” Proc Eur Dial Transplant Assoc. 17:548-556 (1980).
Lambrey, “Possible Link Between Changes in Plasma 24,25-Dihydroxyvitamin D and Healing of Bone Resorption in Dialysis Osteodrstrophy” Metab. Bone Dis. & Rel. Res. 4:25-30 (1982).
Langman et al., “25-Hydroxyvitamin D3 (Calcifediol) Therapy of Juvenile Renal Osteodystrophy: Beneficial Effect on Linear Growth Velocity,” J. Pediatrics, 100:815-820 (1982).
Larrosa M. et al., Long-Term Treatment of Hypovitaminosis D. Calcidol or Cholecalciferol? Annals of the Rheumatic Diseases, vol. 64, No. Suppl. 3, Jul. 2005, p. 366.
Lau et al., “Vitamin D Therapy of Osteoporosis: Plain Vitamin D Therapy Versus Active Vitamin D Analog (D-Hormone) Therapy,” Calcif. Tissue Int., 65:295-306 (1999).
Lehmann et al., “Coating of Tablets and Small Particles with Acrylic Resins by Fluid Bed Technology,” Int. J. Pharm. Tech. & Prod. Mfr., 2:31-43 (1981).
Letteri et al., “Effects of 25-Hydroxycholecalciferol on Calcium Metabolism in Chronic Renal Failure” Adv. Exp. Med. Biol. 81:591-601 (1977).
Lips et al., “A Global Study of Vitamin D Status and Parathyroid Function in Postmenopausal Women with Osteoporosis: Baseline Data from the Multiple Outcomes of Raloxifene Evaluation Clinical Trial,” The Jour. of Clin. Endo. & Meta., 86(3):1212-1221 (2001).
Lomonte et al., “Are Low Plasma Levels of 25-(OH) Vitamin D a Major Risk Factor for Hyperparathyroidism Independent of Calcitriol in Renal Transplant Patients?” J. Nephrol., 18:96-101 (2005).
Lund et al., “Serum 1,25-Dihydroxycholecalciferol in Anephric. Haemodialyzed and Kidney-transplanted Patients,” Nephron, 25:30-33 (1980).
Maierhofer et al., “Synthesis and Metabolic Clearance of 1,25-Dihydroxyvitamin D as Determinants of Serum Concentrations: a Comparison of Two Methods” Journal of Clinical Endocrinology and Metabolism 53:472-475 (1981).
Manni et al., “Oral Calcitriol: Comparison Between the Same Weekly Dose Administered as a Single vs. Two Divided Pulsed Doses in Secondary Hyperparathyroidism of Chronic Renal Failure,” Ital. J Mineral Electrolyte Metab., 11:61-64 (1997).
Martin et al., “19-Nor-1-α-25-Dihydroxyvitamin D2 (Paricalcitol) Safely and Effectively Reduces the Levels of Intact Parathyroid Hormone in Patients on Hemodialysis,” J. Am. Soc. Nephrol., 9:1427-1432 (1998).
Matsushita et al., “Clinical effects of 25-hydroxycholecalciferol in patients with chronic renal failure,” J Nutr Sci Vitaminol, 23:257-261 (1977).
Mazouz et al., “Risk factors of renal failure progression two years prior to dialysis” Clinical Nephrology 51 (6):355-366 (1999).
Mazur, “Effects of 25-OHD3 on Renal Function in Pediatric Patients with Chronic Renal Failure,” Mineral Electrolyte Metab. 10:351-358 (1984).
Memmos et al., “Response of uremic osteoid to vitamin D,” Kidney Int, 21 (Suppl. 11): S50-S54 (1982).
Menon et al., “Vitamin D insufficiency and hyperparathyroidism in children with chronic kidney disease,” Pediatr Nephrol, 23:1831-1836 (2008).
Messa et al., “Direct In Vivo Assessment of Parathyroid Hormone-Calcium Relationship Curve in Renal Patients,” Kidney Int., 46:1713-1720 (1994).
Minutes of US FDA E&M Advisory Committee Meeting of Oct. 4, 1979 for Calderol® calcifediol capsules.
Moe et al., “A randomized trial of cholecalciferol versus doxercalciferol for lowering parathyroid hormone in chronic kidney disease,” Clin.J.Am.Soc.Nephrol. 5: 299-306 (2010).
Moe et al., “Safety and Efficacy of Pulse and Daily Calcitriol in Patients on CAPD: A Randomized Trial,” Nephrol. Dial. Transplant., 13:1234-1241 (1998).
Morris, “Cats Discriminate Between Cholecalciferol and Ergocalciferol,” J. Anim. Physiol, a. Anim. Nutr., 86:229-238 (2002).
Morris, “Vitamin D: A Hormone for All Seasons—How Much is Enough?” Clin. Biochem. Rev., 26:21-32 (2005).
Muindi et al., “Phamacokinetics of Liquid Calcitriol Formulation in Advanced Solid Tumor Patients: Comparison with Caplet Formulation,” Cancer Chemother. Pharmacol., 56:492-496 (2005).
Naik et al., “Effects of Vitamin D Metabolites and Analogues on Renal Function,” Nephron, 28:17-25(1981).
Nakanishi et al., “The Roles of Vitamin D in Secondary Hyperparathyroidism,” [journal in Japanese] 52:1107-1112 (2004).
Norman et al. (eds.), Vitamin D—Gene Regulation, Structure-Function Analysis and Clinical Application: Proceedings of the Eighth Workshop on Vitamin d Paris, France, pp. 765-766, New York: Walter De Gruyter Inc. (1991).
Notice of allowance dated Jul. 10, 2012, in EPO application 08746908.6.
Oksa et al., “Effects of long-term cholecalciferol supplementation on mineral metabolism and calciotropic hormones in chronic kidney disease,” Kidney Blood Press Res., 31: 322-329 (2008).
Parfitt et al., “Calcitriol But No Other Metabolite of Vitamin D is Essential for NormalBone Growth and Development in the Rat,” J. Clin. Invest., 73:576-586 (1984).
Patel et al., “Glomerular Filtration Rate is a Major Determinant of the Relationship Between 25-Hydroxyvitamin D and Parathyroid Hormone,” Calcif. Tissue Int., 80:221-226 (2007).
Peacock et al., “Effect of Calcium or 25OH Vitamin D3 Dietary Supplementation on Bone Loss at the Hip in Men and Women over the Age of 60” The Journal of Clinical Endocrinology & Metabolism, 85(9):3011-3019 (2007).
Perrie, Pharmaceutics: Drug Delivery and Targeting, Second Edition, Chapter 1 (2012).
Phadnis et al., “Direct, Rapid Effects of 25-Hydroxyvitamin D3 on Isolated Intestinal Cells,” J. Cell. Biochem., 90:287-293 (2003).
Pourgholami et al., “1,25-Dihydroxyvitamin D3 Dissolved in Lipiodol Produces a Sustained Antiproliferative Effect in the Human Hepatoblastoma Cell Line HepG2,” Anticancer Res., 20:723-728 (2000).
Pourgholami et al., “In Vitro Antiproliferative Activity of a Medium-Chain Triglyceride Solution of 1, 25-Dihydroxyvitamin D3 in HepG2 Cells,” Anticancer Res., 20:4257-4260 (2000).
Prescribing Information for Calderol® calcifediol capsules (1988).
Prescribing Information for Hectorol® (doxercalciferol capsules), Genzyme (2011).
Prosecution History for U.S. Appl. No. 11/549,001, filed Oct. 12, 2006.
Prosecution History for U.S. Appl. No. 13/244,945, filed Sep. 26, 2011.
Rambeck et al., “Biological Activity of 1 α,25-Dihydroxyergocalciferol in Rachitic Chicks and in Rats,” IZVIAK, 54(2/3):135-139 (1984).
Rapuri, P.B. et al., “Effect of Vitamins D2 and D3 Supplement Use on Serum 25-OHD Concentration in Elderly Women in Summer and Winter,” Calcified Tissue International, 74(2):150-156 (2004).
Recker et al., “The Efficacy of Calcifediol in Renal Osteodystrophy,” Arch. Intern. Med., 138:857-863 (1978).
Reddy et al., Abstracts Sixth Annual Scientific Meeting of the American Society for Bone and Mineral Research, 36:524 (1984).
Reichel et al., “Calcium Metabolism in Early Chronic Renal Failure: Implications for the Pathogenesis of Hyperparathyroidism,” Nephrol. Dial. Transplant., 6:162-169 (1991).
Reichel et al., “Intermittent Versus Continuous Administration of 1,25-dihydroxyvitamin D3 in experimental renal hyperparathyroidism,” Kidney Int., 44:1259-1265 (1993).
Reichel, “Current treatment options in secondary renal hyperparathyroidism,” Nephrol Dial Transplant 21:23-28 (2006).
Ritter et al., “25-Hydroxyvitamin D3 suppresses PTH synthesis and secretion by bovine parathyroid cells,” Kidney Int., 70:654-659 (2006).
Rix et al., “Effect of 18 Months of Treatment with Alfacalcidol on Bone in Patients with Mild to Moderate Chronic Renal Failure,” Nephrol Dial Transplant, 19:870-876 (2004).
Rucker et al., “Vitamin D insufficiency and treatment with oral vitamin D3 in northern-dwelling patients with chronic kidney disease,” J.Nephrol. 22: 75-82 (2009).
Russell et al., “Therapeutic Effects of 25-Hydroxyvitamin D3 on Renal Osteodystrophy,” Mineral Electrolyte Metab., 1:129-138 (1978).
Rutherford et al., “Effect of 25-Hydroxycholecalciferol on Calcium Absorption in Chronic Renal Disease,” Kidney International, 8:320-324 (1975).
Saab et al., “Prevalence of Vitamin D Deficiency and the Safety and Effectiveness of Monthly Ergocalciferol in Hemodialysis Patients,” Nephron Clin. Pract., 105:c132-c138 (2007).
Sanchez, “Prevention and Treatment of Renal Osteodystrophy in Children With Chronic Renal Insufficiency and End-Stage Renal Disease,” Seminars in Nephrology, 21:441-450 (2001).
Schmidt, “Measurement of 25-Hydroxyvitamin D Revisited,” Clinical Chemistry, 52(12):2304-2305 (2006).
Sebert et al. “Comparative effects of equal doses of vitamin D2 and vitamin D3 for the correction of vitamin D deficiency in the elderly” in Norman et al. (eds.), Vitamin D—Gene Regulation, Structure-Function Analysis and Clinical Application: Proceedings of the Eighth Workshop on Vitamin d Paris, France, pp. 765-766, New York: Walter De Gruyter Inc. (1991).
Sebert et al., “Effets A Long Terme D'Une Association De 25-Hydroxycholécalciférol et de 1-Alpha-Hydroxycholécalciférol Sur L'Ostéodystrophie Des Hémodialysés Chroniques” Rev. Rhum Mal Osteoartic 48(7-9):535-541 (1981).
Sebert et al., “Limit by Hyperphosphatemia of the Usefulness of Vitamin D Metabolites (1 alpha-Hydroxycholecalciferol and 25-Hydroxycholecalciferol) in the Treatment of Renal Osteodystrophy,” Metab. Bone Dis. & Rel. Res., 2:217-222 (1980).
Sekkarie, “The Impact of Over-the-counter Vitamin D Suppiementals on Vitamin D and Parathyroid Hormone Levels in Chronic Kidney Disease,” Clin. Nephrology, 65:91-96 (2006).
Shah et al., “Prevalence and correction of 25(OH) vitamin D deficiency in peritoneal dialysis patients,” Peritoneal Dialysis Int., 25:362-366 ( 2005).
Shi et al., “Preparation of Chitosan/Ethylcellulose Complex Microcapsule and its Application in Controlled Release of Vitamin D2,” Biomaterials, 23:4469-4473 (2002).
Singh et al., “C-3 Epimers Can Account for a Significant Proportion of Total Circulating 25-Hydroxyvitamin D in Infants, Complicating Accurate Measurement and interpretation of Vitamin D Status,” J. Clin. Endo. & Metabol., 91(8):3055-3061 (2006).
Sjoden, et al., “1α-Hydroxyvitamin D2 is Less Toxic than 1α-Hydroxyvitamin D3 in the Rat,” Society for Experimental Biology and Medicine, 179: 432-436 (1985).
Skelly et al., In vitro and in vivo testing an correlation for oral controlled/modified-release dosage forms. Pharm. Res., 7(9):975-82 (1990).
Slatopolsky et al., “Differential Effects of 19-nor-1,25-(OH)2D2 and 1 α-Hydroxyvitamin D2 on Calcium and Phosphorus in Normal and Uremic Rats,” Kidney International, 62:1277-1284 (2002).
Somerville et al., “Resistance to Parathyroid Hormone in Renal Failure: Role of Vitamin D Metabolites,” Kidney Int., 14:245-254 (1978).
Sommerfeldt et al., “Metabolism of Orally Administered [3H]Ergocalciferol and [3H]Cholecalciferol by Dairy Calves,” J. Nutr., 113:2595-2600 (1983).
Stamp et al., “Comparison of Oral 25-Hydroxycholecalciferol, Vitamin D, and Ultraviolet Light as Determinants of Circulating 25-Hydroxyvitamin D,” The Lancet, 1341-1343 (Jun. 25, 1977).
Stamp, “Intestinal Absorption of 25-hydroxycholecalciferol,” The Lancet, 121-123 (1974).
Stein et al., “An Update on the Therapeutic Potential of Vitamin D Analogues,” Expert Opin. Investig. Drugs, 12:825-840 (2003).
Stubbs et al., “Cholecalciferol supplementation alters calcitriol-responsive monocyte proteins and decreases inflammatory cytokines in ESRD,” J.Am.Soc.Nephrol., 21: 353-361 (2010).
Stumpf, “The Dose Makes the Medicine,” Drug Discovery Today, 11:550-555 (2006).
Szycher, Szycher's Dictionary of Biomaterials and Medical Devices, pp. 20, 48, 127, Lancaster, Penn: Technomic Publishing Co., Inc. (1992).
Sömjen et al., “Nonhypercalcemic Analogs of Vitamin D Stimulate Creatine Kinase B Activity in Osteoblast-Like ROS 17/2.8 Cells and Up-Regulate Their Responsiveness to Estrogens,” Steroids, 63:340-343 (1998).
Taylor et al., “Interrelationship of Serum 25-Hydroxyvitamin D3 and 1,25-Dihydroxyvitamin D in Juvenile Renal Osteodystrophy after Therapy with 25-Hydroxyvitamin D3,” Metab. Bone Dis. & Rel. Res., 4:255-261 (1982).
Taylor et al., “The absence of 24,25-dihydroxycholecalciferol in anephric patients,” Clin.Sci.Mol.Med.Suppl., 55: 541-547 (1978).
Taylor, CM, 24,25-Dihydroxyvitamin D in Human Serum. In: Vitamin D. Basic Research and Clinical Applications, pp. 197-203. Walter de Gruyter, New York (1979).
Teitelbaum et al., “Calcifediol in Chronic Renal Insufficiency” JAMA 235(2):164-167 (1976).
Teitelbaum et al., “Tetracycline fluorescence in uremic and primary hyperparathyroid bone,” Kidney Int., 12:366-372 (1977).
Thomas et al., “Hypovitaminosis D in Medical Inpatients,” NEJM, 338:777-783 (1998).
Thombre, “Assessment of the feasibility of oral controlled release in an exploratory development setting,” Drug Discovery Today, 10(17): 1159-1166 (2005).
Tokmak et al., “High-dose cholecalciferol to correct vitamin D deficiency in haemodialysis patients,” Nephrol.Dial.Transplant., 23: 4016-4020 (2008).
Trakarnvanich et al., “Effect of high dose ergocalciferol in chronic kidney disease patients with 25-hydroxyvitamin D deficiency,” J.Med.Assoc.Thai. 93: 885-891 (2010).
Tuohimaa et al., “Both High and Low Levels of Blood Vitamin D are Associated with a Higher Prostate Cancer Risk: A Longitudinal, Nested Case-Control Study in the Nordic Countries,” Int. J. Cancer, 108(1):104-108 (2004).
US FDA Clinical Review and Evaluation of NDA for Calderol® calcifediol capsules (believed to be available circa 1983).
US FDA Summary of Basis of Approval for Calderol® calcifediol capsules (believed to be available circa 1980).
Van Weelden et al., “Apoptotic Regression of MCF-7 Xenografts in Nude Mice Treated with the Vitamin D3 Analog, EB1089,” Endocrinology, 139:2102-2110 (1998).
Verberckmoes et al., “Osteodystrophy of Dialysed Patients Treated with Vitamin D,” Proc Eur Dial Transplant Assoc., 10(0): 217-226 (1973).
Vieth, “Vitamin D Supplementation, 25-Hydroxyvitamin D Concentrations, and Safety,” Am. J. Clin. Nutr., 69:842-856 (1999).
Vieth, “What is the optimal vitamin D status for health?” Prog. Biophys. Mol. Biol., 92:26-32 (2006).
Wise (ed.), Handbook of Pharmaceutical Controlled Release Technology, “An Overview of Controlled Release Systems,” Chapter 22, pp. 431-445, 461-463; Research and Development Aspects of Oral Controlled-Release Dosage Forms, Chapter 23, pp. 465-473, New York: Marcel Dekker, Inc. 3 (2000).
Witmer et al., “Effects of 25-hydroxycholecalciferol on bone lesions of children with terminal renal failure” Kidney International 10:395-408 (1976).
Wootton, “Improving the Measurement of 25-Hydroxyvitamin D,” Clin Biochem Rev, 26:33-36 (2005).
Written Opinion for Application No. PCT/US2007/061521, dated Jul. 17, 2007.
Written Opinion for Application No. PCT/US2008/061579, dated Aug. 21, 2008.
Yanoff et al., “The Prevalence of Hypovitaminosis D and Secondary Hyperparathyroidism in Obese Black Americans,” Clin. Endocrinol. (Oxf), 64(5):523-529 (2006).
Prescribing Information forZemplar® (paricalcitol) Capsules, Abbott (2011).
Zerwekh et al., “Extra-Renal Production of 24,25-Dihydroxyvitamin D in Chronic Renal Failure During 25 Hydroxyvitamin D3 Therapy,” Kidney Int., 23:401-406 (1983).
Zisman et al., “Impact of Ergocalciferol Treatment of Vitamin D Deficiency on Serum Parathyroid Hormone Concentrations in Chronic Kidney Disease,” Am. J. Nephrol., 27:36-43 (2007).
Zucchelli et al., “Therapeutic effects of 25-hydroxycholecalciferol and sodium etidronate on renal osteodystrophy,” Mineral.Electrolyte Metab. 7: 86-96 (1982).
AlfaD3® 0.25, 0.5 or 1 microgram Capsules (Alfacalcidol, Package Leaflet, Apr. 2010).
Alfarol® Capsules 3 μg (Package Leaflet, Mar. 2011).
Ashford, Chapter 20: Bioavailability—physicochemical and dosage form factors, pp. 314-333 In: Aulton et al. (eds.), Aulton's Pharmaceutics. The Design and Manufacture of Medicines, Fourth Edition, Elsevier Publishing (2013).
Hectorol® (doxercalciferol) Capsules (Label, FDA, 2010).
Zemplar® (paricalcitol) Capsules, Final Agreed Upon Label (FDA, May 5, 2009).
Sitrin et al., Comparison of vitamin D and 25-hydroxyvitamin D absorption in the rat, Am. J. Physiol., 242(4):G326-32 (1982).
Holmberg et al., Absorption of a pharmacological dose of vitamin D3 from two different lipid vehicles in man: comparison of peanut oil and a medium chain triglyceride, Biopharm. Drug Dispos., 11(9):807-15 (1990).
9 Things That Can Undermine Your Vitamin D Level: Don't Let Your Vitamin D Absorption Slip Away, Harvard Health Publishing, downloaded from the Internet at: <https://www.health.harvard.edu/healthbeat/9-things-that-can-undermine-your-vitamin-d-level> (Feb. 11, 2019)., Feb. 11, 2019.
ACP Formulary and Pocket Guide to Psychopharmacology, Virginia DMHMRSAS, vol. 1, Iss. 1 (2004-2005).
Albertson et al., Quantitative mapping of amplicon structure by array CGH identifies CYP24 as a candidate oncogene, Nat. Genet., 25(2):144-6 (2000).
Amin, The impact of improved phosphorus control: use of sevelamer hydrochloride in patients with chronic renal failure, Nephrol Dial Transplant, 17:340-345 (2002).
Anderson et al., Expression of VDR and CYP24A1 mRNA in human tumors, Cancer Chemother. Pharmacol., 57(2):234-40 (2006).
Anderson et al., Quantification of mRNA for the vitamin D metabolizing enzymes CYP27B1 and CYP24 and vitamin D receptor in kidney using real-time reverse transcriptase-polymerase chain reaction, 2003. J. Mol. Endoc 31:123-132.
Baggiolini et al., “Stereocontrolled Total Synthesis of 1 alpha, 25-Dihydroxycholecalciferol 1 and 1 alpha, 25-Dihydroxyergocalciferol,” J Org Chem. 21: 3098-3108 (1986).
Berg et al., 24,25-Dihydroxyvitamin d3 and vitamin D status of community-dwelling black and white Americans, Clin. Chem., 61(6):877-84 (Jun. 2015).
Berruti et al., Prognostic role of serum parathyroid hormone levels in advanced prostate cancer patients undergoing zoledronic acid administration, Oncologist, 17(5):645-52 (2012).
Bertoldo et al., Serum 25-hydroxyvitamin D levels modulate the acute-phase response associated with the first nitrogen containing bisphosphonate infusion, J. Bone Miner. Res., 25(3):447-54 (Mar. 2010).
Bhatia et al., EB1089 inhibits the parathyroid hormone-related protein-enhanced bone metastasis and xenograft growth of human prostate cancer cells, Mol. Cancer Ther., 8(7):1787-98 (2009).
BioTrends Research Group, TreatmentTrends (Registered): Nephrology (US) Q4 2014 (Dec. 2014).
Blunt et al., Biological Activity of 25-Hydroxycholecalciferol, A Metabolite of Vitamin D3, Proc. N.A.S., USA, 61(4):1503-6 (1968).
Boxtel et al., “Drug Benefits and Risks, International Textbook of Clinical Pharmacology,” p. 75-76 (2001).
Briese et al., “Arterial and cardiac disease in young adults with childhood-onset end-stage renal disease-impact of calcium and vitamin D therapy,” Nephrology Dialysis Transplantation., 21:1906-1914 (2006).
Brodowicz et al., Early identification and intervention matters: A comprehensive review of current evidence and recommendations for the monitoring of bone health in patients with cancer, Cancer Treat Rev., 61:23-34 (2017).
Centorrino et al., “Multiple versus single antipsychotic agents for hospitalized psychiatric patients: case-control study of risks versus benefits,” Am J. Psychiatry, 161(4): 700-06 (2004).
Charnow, Novel Formulation Corrects Vitamin D, Lowers iPTH, Renal & Urology News (2012).
Chen et al., Safety of Denosumab Versus Zoledronic Acid in Patients with Bone Metastases: A Meta-Analysis of Randomized Controlled Trials, Oncol. Res. Treat., 39(7-9):453-9 (2016).
Chonchol et al., 25-Hydroxyvitamin D, insulin resistance, and kidney function in the Third National Health and Nutrition Examination Survey, Kidney Int., 71(2):134-9 (2007).
Database WPI Week 199546 Thomson Scientific, London, GB; AN 1995-355178 XP002464406.
Database WPI Week 199546 Thomson Scientific, London, GB; AN 1995-355178 XP002680886.
E.W. Martin, “Drug Interactions,” in Hazards of Medication, J.B. Lippincott Co. (1978).
El Abdaimi et al., Reversal of hypercalcemia with the vitamin D analogue EB1089 in a human model of squamous cancer, Cancer Res., 59(14):3325-8 (1999).
Ennis et al., Current recommended 25-hydroxyvitamin D targets for chronic kidney disease management may be too low, J. Nephrol., 29(1):63-70 (Feb. 2016).
Epps et al., “Vitamin D Metabolism: Implications for Treatment in Oncology,” Oncology News, 4:42-44 (2009).
Final Office Action, U.S. Appl. No. 16/089,235, dated Dec. 10, 2019, 35 pages.
Fliser et al., Fibroblast gowth factor 23 (FGF23) predicts progression of chronic kidney disease: the mild to moderate kidney disease (MMKD) study, J. Am. Soc. Nephrol, 18:2601-8 (2007).
Fournier et al., “Impact of calcium and vitamin D therapy on arterial and cardiac disease in young adults with childhood-onset and stage renal disease,” Nephrol Dial Transplant, 22:956-957 (2006).
Friedrich et al., Analysis of the vitamin D system in cervical carcinomas, breast cancer and ovarian cancer, Recent Results Cancer Res., 164:239-46 (2003).
Fukagawa et al., FGF23: its role in renal bone disease, Pediatr. Nephrol., 21:1802-6 (2006).
Fukagawa et al., With or without the kidney: the role of FGF23 in CKD, Nephrol. Dial. Transplant., 20:1295-8 (2005).
Gal-Moscovici et al., Role of vitamin D deficiency in chronic kidney disease, Journal of Bone and Mineral Res. 22:V91-V94 (2007).
Garland et al., Vitamin D for cancer prevention: global perspective, Ann. Epidemiol., 19(7):468-83 (2009).
Goldzieher JW(Endocr Pract. Sep.-Oct. 1999;5(5):229-32).
Gopinath et al., Disintegrants—A Brief Review, J. Chem. Pharm. Sci., 5(3):105-12 (Jul.-Sep. 2012).
Guidance for Industry, Nonclinical Safety Evaluation of Drug or Biologic Combinations, U.S. Department of Health and Human Services, Food and Drug Administration (Mar. 2006).
Haddad et al., “Vitamin D Plasma Binding Protein. Turnover and Fate in the Rabbit,” J. Clin. Invest., 67(5): 1550-1560 (1981).
Harris R Z et al: “Pharmacokinetics of cinacalcet hydrochloride when administered with ketoconazole”, Clinical Pharmacokinetics, Adis International Ltd., Auckland, NZ, vol. 46, No. 6, Jan. 1, 2007 (Jan. 1, 2007), pp. 495-501.
Helvig et al., Dysregulation of renal vitamin D metabolism in the uremic rat, Kidney Int., 78(5):463-72 (2010).
Hemodialysis (2015, 4 pages, Accessed from https://www.kidney.org/atoz/content/hemodialysis on Jun. 19, 2019) (Year: 2015).
Henry et al., Response of chick parathyroid glands to the vitamin D metabolites, 1,25-dihydroxycholecalciferol and 24,25-dihydroxycholecalciferol, J. Nutr., 107(10):1918-26 (1977).
Holick et al., “Vitamin D2 is as effective as vitamin D3 in maintaining circulating concentrations of 25-dydroxyvitamin D,” J Clin Endocrinol Metab., 93(3):677-81 (2008).
Holick et al., Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline, J. Clin. Endocrinol. Metab., 96(7):1911-30 (Jul. 2011).
Holick, Vitamin D for health and in chronic kidney disease, Semin. Dial., 18(4):266-75 (2005).
Ibrahim et al., Serum fibroblast growth factor-23 levels in chronic haemodialysis patients, Int. Urol. Nephrol., 41:163-9 (2009).
Inoue et al., Role of the vitamin D receptor in FGF23 action on phosphate metabolism, Biochem. J., 399:325-31 (2005).
International Preliminary Report on Patentability for Corresponding International Application No. PCT/EP17/57282, dated Oct. 11, 2018, 7 pages.
International Preliminary Report on Patentability for Corresponding International Application No. PCT/US09/39355, dated Oct. 14, 2010, 8 pages.
Jones et al., Cytochrome P450-mediated metabolism of vitamin D, J. Lipid Res., 55(1):13-31 (2014).
Wagner et al., The ratio of serum 24,25-dihydroxyvitamin D(3) to 25-hydroxyvitamin D(3) is predictive of 25-hydroxyvitamin D(3) response to vitamin D(3) supplementation, J. Steriod Biochem. Mol. Biol., 126(3-5):72-7 (Sep. 2011).
Wang-Gillam et al., Evaluation of vitamin D deficiency in breast cancer patients on bisphosphonates, Jul. 1, 2008, Oncologist, 821-7, 13(7).
Yueh-Ting et al: Comparison between Calcitriol and Caltiriol Plus Low-Dose Cinacalcet for the Treatment of Moderate to Severe Secondary Hyperparathyroidism in NUTRIENTS, vol. 5, No. 4, Apr. 19, 2013 (Apr. 19, 2013), pp. 1336-1348.
Zerwekh J. E.: “Blood biomarkers of vitamin D status”, The American Journal of Clinical Nutrition, vol. 87Suppl., 2008, pp. 1087S-1091S.
Zuradelli et al., High incidence of hypocalcemia and serum creatinine increase in patients with bone metastases treated with zoledronic acid, Oncologist, 14(5):548-56 (2009).
Hirata et al., “A comparison between 1,25-dihydroxy-22-oxavitamin D3 and 1,25-dihydroxyvitamin D3 regarding suppression of parathyroid hormone secretion and calcaemic action,” Nephrol Dial Transplant 17:41-45 (2002).
Meyer et al., “A kidney-specific genetic control module in mice governs endocrine regulation of the cytochrome P450 gene Cyp27b1 essential for vitamin D3 activation,” J. Biol. Chern. 292(42):17541-17558 (2017).
Petkovich et al., “Modified-release oral caldifediol corrects vitamin D insufficiency with minimal CYP24A1 upregulation,” Journal of Steroid Biochemistry & Molecular Biology 148:283-289 (2015).
Segersten et al., “25-Hydroxyvitamin D3-1α-Hydroxylase Expression in Normal and Pathological Parathyroid Glands,” J Clin Endocrinol Metab, 87(6):2967-2972 (2002).
Silver et al., “Regulation of PTH synthesis and secretion relevant to the management of secondary hyperparathyroidism in chronic kidney disease,” Kidney International 67(95):S8-S12 (2005).
Sprague et al., “Paricalcitol versus calcitriol in the treatment of secondary hyperparathyroidism,” Kidney International 63:1483-1490 (2003).
Haddad, et al., Acute Administration of 25-Hydroxycholecalciferol in Man, JCE & M, 42 (2): 284-290 (1976).
Sosa et al., The effect of 25-dihydroxyvitamin D on the bone mineral metabolism of elderly women with hip fracture, Rheumatology, 39: 1263-1268 (2000).
Cavalli et al., Biological effects of various regimes of 25-hydroxyvitamin D3 (calcidiol) administration on bone mineral metabolism in postmenopausal women, Clinical Cases in Mineral and Bone Metabolism, 6(2): 169-173 (2009).
Related Publications (1)
Number Date Country
20130137663 A1 May 2013 US
Provisional Applications (2)
Number Date Country
61405196 Oct 2010 US
61318741 Mar 2010 US