Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Jan 27.
Published in final edited form as: Best Pract Res Clin Endocrinol Metab. 2011 Aug;25(4):605–615. doi: 10.1016/j.beem.2011.06.006

Vitamin D and cancer: Clinical aspects

Anna Woloszynska-Read a,b, Candace S Johnson b, Donald L Trump c,*
PMCID: PMC4729360  NIHMSID: NIHMS738348  PMID: 21872802

Abstract

There are substantial preclinical and epidemiologic data that suggest that vitamin D plays a role in the prevention and treatment of cancer. Numerous observational studies have shown that low blood levels of 25(OH) vitamin D (cholecalciferol), estimated by geographical location, diet and activity assessment or measured serum levels are associated with a higher risk of cancer and worse cancer-specific survival as well as numerous morbidities to e.g. cardiovascular disease, stroke, infection, autoimmune disease, and neuromuscular dysfunction among large populations. A considerable number of in vitro and in vivo studies indicate that the most active metabolite of vitamin D – 1,25-dihydroxycholecalciferol or calcitriol – has anti-proliferative, pro-apoptotic, pro-differentiating, and anti-angiogenic properties. Combined treatment of calcitriol and many types of cytotoxic agents has synergistic or at least additive effects. However, clinical trials testing these hypotheses have been less encouraging, though a number of methodological, pharmacological, and pharmaceutical issues confound all trials ever conducted. In order to properly assess the clinical value of vitamin D, its metabolites and analogs in cancer prevention and treatment, more studies are needed.

Keywords: calcitriol, vitamin D, clinical trials

Introduction

In recent years a large number of observational and experimental studies have been undertaken to investigate the role of vitamin D and its metabolites in cancer.14 Vitamin D is a hormone produced through a multistep process initiated in the skin with conversion of 7-dehydrocholesterol to cholecalciferol (D3) upon exposure to ultraviolet B radiation. When cholecalciferol, or the plant derived derivative ergocalciferol, enters the circulation through epidermal transfer or after ingestion and intestinal absorption, it associates with vitamin D-binding protein. In this form it travels to the liver where it is hydroxylated to form 25(OH) vitamin D, the most abundant circulating form of vitamin D. The conversion of 25(OH) vitamin D to 1,25-dihydroxyvitamin D (1,25(OH)2D or calcitriol) occurs predominantly in the kidneys, but other normal and neoplastic tissues are known to express 1-α-hydroxylase, suggesting that this enzymatic reaction also takes place in other sites in the body. Calcitriol together with parathyroid hormone (PTH) maintains proper levels of calcium in the blood, and if calcium is not available from dietary sources, normal levels are maintained at the expense of bone loss.5 In addition to these bone and mineral effects,6 vitamin D compounds have anticancer properties.717 For the purposes of this article we will focus primarily on the clinical applications of vitamin D and its metabolites in cancer treatment.

Although many foods in the US are fortified with vitamin D, the main source of the nutrient comes from its synthesis in the skin upon exposure to the ultraviolet B radiation from the sun.1820 A National Health and Nutrition Evaluation Survey (NHANES) has estimated that an average individual’s food intake of vitamin D in the United States is 200 IU/day; the recently updated recommended dietary allowance by the Institute of Medicine is 600 IU/day for most healthy adults and 800 IU/day for those over the age of 70.2124 The definition of the “correct” levels and the role or necessity for supplementation of vitamin D to improve health and prevent disease are controversial. While some researchers claim that vitamin D deficiency is overestimated, others believe that most of the US population does not get enough of the nutrient from sun exposure and diet. Dialog in health and research communities has become so heated that it prompted the Agency for Healthcare Research and Quality (AHRQ) to charge the Institute of Medicine with the task of revisiting previous recommendations. As a consequence, an IOM study resulted in a new set of recommendations that were issued in the consensus study, “Dietary Reference Intakes for Calcium and Vitamin D,” in late 2010.21 Although the IOM committee concluded that most North Americans are getting enough vitamin D, the new study recommended a daily increase of vitamin D for some groups compared to the recommendations established in a previous IOM report from 1997.21 The report also noted that in contrast to the large body of literature conclusively indicating an important role of vitamin D in skeletal health, the use of vitamin D in cancer chemoprevention and treatment remains ambiguous and inconclusive.21 The ambiguity arises from two aspects of the potential clinical utility of vitamin D that have to be evaluated independently of one another. First, preclinical data show that calcitriol elicits its anti-tumor properties at high systemic concentrations. Second, there is no agreed-upon definition of what levels of 25(OH) vitamin D in healthy individuals are adequate to protect against cancer.25

The most widely used indicator of vitamin D status in the body is 25 hydroxyvitamin D3, a precursor of calcitriol.5 The longer half-life of 25(OH) vitamin D (~ 3 weeks) makes this metabolite the most suitable surrogate marker of body vitamin D stores when compared to other vitamin D metabolites.5 25(OH) vitamin D levels are usually determined by measurement of the metabolite in the plasma or serum. It is important to note, however, that the levels of 25(OH) vitamin D which are ideal or appropriate are not known. The levels constituting deficiency or insufficiency are somewhat arbitrarily based on associations of 25(OH)D3 levels and bone mineral density (BMD) or PTH hormone levels. Using these measures it is generally agreed that levels of 25(OH)D3 below or equal to 20 ng/ml and below or equal to 30 ng/ml represent vitamin D deficiency and insufficiency, respectively. The recommended form for replacement therapy is vitamin D3 (cholecalciferol), since it seems to be more potent than D2 (ergocalciferol)5 and D3 is detectable by all commercial assays.

The 2010 IOM consensus analysis noted that the majority of data regarding the impact of vitamin D on extraskeletal health outcomes, including cancer are inconsistent in several respects.21 First, only individuals with levels of 25(OH) vitamin D below 12 ng/ml are considered vitamin D deficient, and 20 ng/ml found to be the level that is needed for good bone health for practically all healthy individuals(21). Second, multiple factors such as diet, sun exposure, adipose tissue content, and skin pigmentation play critical roles in an individual’s 25(OH) vitamin D systemic levels.26 Last, the IOM report that recommended 4000 IU daily be the upper safe level of supplementation for large unmonitored populations.21

Epidemiologic link

There is considerable controversy regarding the role of vitamin D in cancer risk, incidence, and mortality as there are numerous conflicting data. The first study suggesting an association between vitamin D and cancer incidence or outcome was the seminal study of Garland and Garland.27 They noted increased colon cancer mortality in populations residing in the northeastern United States where there is less exposure to solar radiation throughout the year, and higher mortality among people living in the southern, southwestern, and western portions of the US. These findings led the Garland brothers to suggest that greater UV light exposure led to increased systemic levels of vitamin D and that vitamin D may decrease the risk of colon cancer death. Numerous observational studies followed up on this concept and found associations between geographic distribution and vitamin D serum levels, and geographic site of residence and cancer incidence and mortality.2832 A clear and unequivocal causal link between geographic location, UV irradiation, vitamin D serum content and cancer incidence or outcome has never been established, however. Although several epidemiologic studies demonstrated increased risk of colorectal, prostate, and breast cancers with low serum vitamin D levels, it is not known if the link is causative or associative. A series of studies regarding this issue was published in 2010 by the Cohort Consortium Vitamin D Pooling Project of Rarer Cancers (VDPP), involving 10 cohorts (2588 female and 2135 male) to address the association between vitamin D and cancer.3,3338 The VDPP examined the associations between serum or plasma 25(OH) vitamin D concentrations and the development of 7 types of rarer cancer: endometrial, esophageal, gastric, kidney, non-Hodgkin lymphoma, ovarian, and pancreatic cancers. Those studies failed to find a reduced risk of cancer incidence associated with higher levels > 30 ng/ml of serum 25(OH) vitamin D for these several types of cancers. In contrast, a study conducted using Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study cohort, showed that men with lower 25(OH) vitamin D serum concentrations were at increased risk of bladder cancer compared with men with higher serum levels.39 This observation is consistent with preclinical data in bladder cancer suggesting that exposure to vitamin D (calcitriol) has anticancer properties.40 Furthermore, higher serum 25(OH) vitamin D may be associated with higher concentration of vitamin D metabolites in the bladder.41,42 Consequently, increased exposure of the bladder tissue to these metabolites could promote apoptosis and differentiation and, therefore, reduce neoplastic processes in the bladder epithelium.

Vitamin D and cancer

There is evidence dating back to preclinical studies in the 1980s indicating that vitamin D compounds have anticancer activities.4345 Since these initial studies, substantial preclinical evidence from in vitro and animal model studies suggest that a biologically active metabolites or analogs of vitamin D may play a critical role in cancer etiology and progression through anti-proliferative, pro-apoptotic, pro-differentiating, and anti-angiogenic mechanisms.5,46,47 A number of clinical studies have attempted to assess the efficacy of vitamin D, and its metabolites and analogs in the prevention and treatment of various malignancies including breast, prostate, and liver cancer, among others. Clinical studies have used vitamin D3 and calcitriol alone or in combination with cytotoxic agents. Most clinical trials involving vitamin D and its metabolites have been conducted in prostate cancer patients.4860

The biologic actions of vitamin D are attributed to its most active metabolite, calcitriol. Calcitriol was approved by the FDA for the treatment of chronic kidney disease, or hypoparathyroidism in patients with chronically low calcium levels. Since then, properties of calcitriol have been assessed in clinical studies of patients with different malignancies. Most preclinical data suggest that the optimal anti-tumor effect of calcitriol and other analogs is seen with the administration of high dose calcitriol on an intermittent schedule. Two pharmaceutical formulations of calcitriol are available – oral (Rocaltrol®, Roche Pharmaceutical Corporation) and intravenous (Calcijex®, Abbott Laboratories) – though neither is fully suited for anticancer treatment. Muindi et al have shown that Rocaltrol® was unsuitable for high dose administration because of inconvenience (maximum tolerated dose was not achieved even at doses that required patients to take ~ 80 caplets daily for 3 days each week) and lack of proportional increase in the serum levels and systemic exposure.61 Similar findings have also been reported by Beer and colleagues who observed “saturable absorption” at doses above 0.48 μg/kg.62

A small number of single agent trials utilizing vitamin D3 and 1,25 hydroxyvitamin D have been conducted with limited success. The first clinical studies to evaluate the anti-proliferative and pro-differentiating properties of calcitriol were conducted in the early 1990’s in myelodysplastic syndrome (MDS) and acute leukemia (AML) patients.6366 A regimen of cytarabine and daily calcitriol prolonged remission in elderly patients with both diseases. Although some positive responses were observed in these trials, they were also associated with hypercalcemia in 10–30% of patients. Thus, early attempts at translating experimental vitamin D data to the clinic were not entirely successful, most likely due to the fact that the calcitriol dose employed was optimized for renal osteodystrophy and osteoporosis patients, and the treatment was not evaluated from the standpoint of the anticancer activity of the agent. Nonetheless, these early clinical findings precipitated development of vitamin D analogs with modified chemical structures, in the hope of alleviating hypercalcemia and increasing the anticancer properties of calcitriol.67,68

Vitamin D3 (cholecalciferol) in clinical trials

The observation that 1 alpha hydroxylase (the enzyme involved in hydroxylation of the calcitriol precursor) is present in malignant and normal tissues suggests that most cells in the body are able to synthesize calcitriol if levels of circulating 25 hydorxyvitamin D (cholecalciferol) are high enough.6972 This has led to trials of vitamin D3 in healthy and cancer patients to prevent and treat cancer, respectively. A Women’s Health Initiative trial of 400 IU of vitamin D and calcium, primarily to prevent fractures, showed that women concurrently treated with estrogen therapy had a decreased risk of colorectal cancer.73 Although cancer was not a primary endpoint, this observation supports cancer protective properties of vitamin D even at small doses. In another study, conducted in prostate cancer patients, Woo et al investigated the effect of cholecalciferol on PSA levels and the rate of rise of PSA in these patients. Fifteen patients were treated with 2000 IU of vitamin D3 per day and monitored every 2–3 months. The PSA levels stabilized or decreased in nine patients for 21 months. Overall, PSA doubling time increased from 14.3 months to 25 months in patients treated with cholecaciferol and no side effects were reported.74

To investigate the role of cholecalciferol in metastatic breast cancer patients Amir et al used a high dose of 10 000 IU of cholecalciferol per day for four months.75 Palliative responses were assessed in 40 patients and a significant decrease in the sites of pain was recorded. However, there also was a significant increase in circulating calcium. Although patients did not experience cholecalciferol direct toxicity, no objective responses and clear increase in survival of this regimen was noted.

Calcitriol – clinical trials – single agent studies

Calcitriol has been used in a number of clinical trials, both as a single drug and in combination with chemotherapeutic agents. Based on data from preclinical studies, the anticancer activity of calcitriol requires supraphysiological doses and systemic exposure and before or simultaneous with cytotoxic drug administration. This requires careful development of appropriate dose/schedules of calcitriol and chemotherapy, as well as definition of toxicity and the maximum tolerated dose (MTD) or optimal biologic dose (OBD).

Based on laboratory data, high concentrations of calcitriol that elicit anti-tumor actions are safe and rarely cause limiting toxicity; the safety of intermittent very high dose calcitriol has been confirmed in cancer patients.10,11,15,56 No grade 3 or 4 toxicities have been seen in cancer patients when high dose calcitriol is administered intermittently. However, the current available formulations of calcitriol are not ideal for anti-tumor treatment since it is difficult to achieve systemic stable levels of the drug.56,62 After initial clinical trials in leukemic patients, calcitriol has been thought of as a toxic agent that induces unmanageable hypercalcemia, which has proven to be another impediment to its clinical application. However, this was not really the case, even in the trials in leukemia. Mild – moderate, rapidly reversible hypercalcemia has been seen with high dose intermittent as well as continuous calcitriol regimens. However, clinical consequences of these changes in calcium level have not been seen. Oral administration of calcitriol on a daily schedule in prostate cancer patients proved that the agent could safely be administrated for several months (up to 15 months) without causing dose-limiting hypercalcemia.49

A phase I trial of calcitriol in patients with advanced malignancies was one of the first clinical studies conducted to determine the MTD of calcitriol as a single agent. Thirty-six patients were entered at doses ranging from 2 to 10 μg.76 Calcitriol was administered by subcutaneous injection every other day. Pharmacokinetic evaluation revealed that serum calcitriol levels were maintained at near peak concentrations for at least 8 h following injection. This study showed that substantial doses of calcitriol can be administered via this route with tolerable toxicity. Another phase I study conducted by Beer and colleagues used a dose-escalating regimen of calcitriol. The agent was administered orally once per week for twenty cycles and no limiting toxicity was observed at doses up to 2.8 μg per kilogram weekly. However, the trial was terminated when incomplete and unreliable oral absorption at the dose 2.4 μg per kilogram and higher became apparent.62

Beer et al treated 22 patients for a median of ten months with 0.5 μg/kg of calcitriol.48 Their study showed that a weekly high dose did not cause significant toxicity and was safe in patients with a rising PSA after prostatectomy. Although the primary endpoint of the study, 50% reduction in the serum PSA, was not achieved, the study did show that a weekly regimen is effective in achieving plasma concentrations > 1 nM; this concentration, based on in vivo studies is higher than the minimum needed for calcitriol anti-tumor activity. Furthermore, this study proved that long-term calcitriol therapy was well tolerated by patients and did not cause limiting (> grade three) toxicity.

When 2–2.5 μg of calcitriol was given to prostate cancer patients daily there was a decrease in the rise of serum PSA or stabilization of PSA levels in those patients, suggesting slower progression of the disease upon calcitriol exposure.49 However, urinary calcium increased and the authors were concerned that the risk of developing renal stones may be higher, and could outweigh the modest benefits of the calcitriol treatment.49 To reduce the risk of hypercalciuria and avert renal toxicity and also potentially to achieve better efficacy of calcitriol, some groups employed intermittent administration of very high doses once or three times a week.48,50,77 This approach caused transient and short-term hypercalcemia, there was no clearly increased risk of renal stones or any other limiting toxicity. Substantially higher serum concentrations of calcitriol were also recorded, which would be expected to increase the potential for anti-tumor effects.

Although oral administration of high doses of calcitriol is safe, our group and that of Beer have shown that the pharmacokinetics of the commercially available oral formulations does not lead to a proportional increase in serum levels or predictable and maximal systemic exposure.56,61 This formulation is not therefore suitable for achieving high enough serum concentrations to elicit anticancer properties. Following these findings an improved oral formulation of calcitriol – DN-101 – was developed by Novacea Pharmaceuticals. A phase I study in patients with advanced malignancies was conducted to assess the safety of the new formulation of calcitriol.52,78 Pharmacokinetics of DN-101 showed that there was a linear relationship between the dose and AUC. A dose of 165 mcg on week one followed by 45 mcg did not produce limiting toxicity. This formulation was then developed in men with prostate cancer in combination with docetaxel.

Calcitriol combination studies

Preclinical studies in cell and animal models have shown that calcitriol can interact in either a synergistic or additive manner with other chemotherapeutics.7983 These findings support the development of combination therapy trials. Phase I and phase II studies involving calcitriol with docetaxel, gefitinib, paclitaxel, and carboplatin in advanced malignancies patients have been completed.54,55,84,85 Beer et al. conducted a combination calcitriol and docetaxel phase I and II clinical trials in patients with castration-resistant prostate cancer. The study evaluated weekly administration of 0.5 μg/kg calcitriol orally on day one followed by 36 mg/m2 docetaxel intravenously on day two, and the cycles were repeated for six consecutive weeks. Five patients who had completed all calcitriol/docetaxel cycles achieved PSA reductions of > 50%. The treatment was well tolerated and only one patient experienced reversible, grade three toxicity.86 Similar promising results were seen in another phase II trial, conducted by the same group, in a larger cohort of prostate cancer patients (37 patients). Beer et al sought to determine the safety and efficacy of weekly high dose oral calcitriol (Rocaltrol®) and docetaxel in patients with metastatic prostate cancer.85 The group found that the combination of weekly oral high dose calcitriol and weekly docetaxel is a well-tolerated regimen for prostate cancer patients. Furthermore, the anti-tumor effect as assessed by ≥50% reduction in PSA, was substantially higher (81%) than would have been expected with weekly docetaxel alone.85 However, the calcitriol formulation (Rocaltrol®) that was used in these studies is pharmaceutically and pharmocokinetically unattractive, as noted above.

Non-cytotoxic drugs have also been used in combination with calcitriol. Glucocorticoids such as dexamethasone ameliorate calcitriol-induced hypercalcemia which allows for a higher dose regimen.53 Additionally, in preclinical studies, dexamethasone substantially enhances calcitriol anti-tumor activity. To examine this combination, Trump and colleagues conducted a phase II study of intravenous calcitriol at a dose of 74 μg weekly and dexamethasone in patients with castration-resistant prostate cancer.53 While anti-tumor activity was noted as evidenced by PSA decreases in and long-term progression free survival of up to 3 years was seen, the level of activity in unselected patients was not sufficient to justify further exploration of this regimen. Calcitriol enhances the anti-tumor activities of several cytotoxic or differentiating agents. Fakih and colleagues conducted a phase I clinical study to determine the MTD of intravenously administered calcitriol in combination with dexamethasone and the EGFR antagonist, gefitinib (Iressa®).55 Although the primary aim of the study was to establish the MTD of this combination treatment, a secondary objective was to examine the pharmacokinetic profile of intravenous calcitriol. The MTD was 125 μg/week calcitriol and the study found that the mean peak serum calcitriol concentration was 11.17 ng/ml and AUC was 54.89 ng h/ml; these are concentrations that are associated with anti-tumor activity in in vivo studies. In a second stage of this trial, simultaneously administered dexamethasone increased the MTD of calcitriol (120 mcg weekly).

These preclinical studies of combinations of cytotoxics and calcitriol provide support for the continued investigation of such combinations. Platinum analogs, taxanes, and DNA-intercalating agents are all potentiated by calcitriol, but only when administrated before or during cytotoxic treatment.87,88

ASCENT-1 trial

The commercially available calcitriol formulations are not suitable for use in cancer chemotherapy. A new oral formulation of calcitriol for cancer treatment was developed by Novacea. DN-101 was an oral high concentration formulation of calcitriol that was available in 15 and 45 μg caplets. This formulation demonstrated linear pharmacokinetics and dose-proportional pharmacokinetic relationship between Cmax and AUC at doses up to 165 μg. DN-101 safety and pharmacokinetics were evaluated in a phase I clinical study.52,78 Based on the development of grade 2 hypercalcemia in two out of seven patients, the phase I study of DN-101 declared the maximal tolerated dose to be 60 μg per week. Subsequently, in what was called a phase II trial, a placebo-controlled study called ASCENT-1, was conducted which tested weekly calcitriol (45 μg) or placebo in combination treatment with docetaxel (36 mg/m2 weekly for 3 out of 4 weeks) in prostate cancer patients with disease progressing despite androgen deprivation,53 250 men with metastatic, castration-resistant prostate cancer were randomly assigned to the placebo + docetaxel or calcitriol + docetaxel. The primary endpoint of the study was frequency of a 50% reduction of PSA within six months of starting the treatment. While PSA response was not significantly different, placebo (52%) vs. calcitriol (63%), patients in the DN-101 arm (calcitriol) had improved survival (24.5 months) when compared to the placebo arm (16.4 months). In addition to improved survival in the DN-101 and docetaxel group, adverse events were reduced. While this positive clinical outcome was very encouraging, this trial was not designed to delineate a difference in survival between these two treatments. No difference in PSA response was noted. A larger phase III study to determine if survival was improved was initiated.60

ASCENT-2 trial

The randomized ASCENT II trial was conducted using the same high dose of calcitriol in combination with docetaxel in patients with progressive castration-resistant prostate cancer.60 The primary objective of the trial was duration of survival; secondary endpoints included rates of thromboembolic events, gastrointestinal events, and overall serious adverse events, all events that seemed reduced in ASCENT-1. Patients were randomly assigned in a double blind fashion to two arms of the study. The experimental arm was identical to that employed in ASCENT 1(28-day dosing cycle of 45 μg DN-101 on days 1, 8, and 15; 36 mg/m2 docetaxel on days 2,9, and 16; and 8 mg oral dexamethasone). However, when the ASCENT-2 trial was initiated the FDA-approved standard regimen for docetaxel in men with castration-resistant prostate cancer was 75 mg/m2 administered every-3-weeks. In a prospective randomized trial docetaxel 75 mg/m2 every 3 weeks was superior to 36 mg/m2 weekly 3 weeks out of 4 [median survival 18.9mo vs. 17.4mo, respectively].89 Therefore this standard therapy of every-3-weeks was chosen as the control arm of ASCENT-2 combined with placebo and was compared to weekly docetaxel plus calcitriol.

Interim analysis by the Data Safety Monitoring Board for this study showed inferior survival in the experimental arm and the study was halted. At the point of terminating the study there were 953 patients enrolled in the trial.60 Median overall survival in the ASCENT arm was 17.8 months and 20.2 months in the control arm (P = .002).

The inferior survival in the calcitriol arm was surprising. While this result may stem from the lack of activity of calcitriol in potentiating docetaxel in men with castration-resistant prostate cancer at least two other concerns must be considered:

  1. Since weekly docetaxel has been proven to be inferior to every-3-weeks docetaxel, the failure of the experimental arm in this study was clearly confounded by the inferiority of the docetaxel regimen.

  2. Another factor that must be considered in evaluating this study is the lack of convincing data that this calcitriol dose and regimen is optimal or appropriate. The dose employed was ~ 1/3 of the MTD of intermittent calcitriol. Therefore, one must wonder whether the failure of this trial was influenced by the dose of calcitriol employed.

Notably, both arms had the same rates of total adverse effects, but the number of deaths was significantly higher in the ASCENT arm (17%) when compared to the control arm (10.1%). The majority of early deaths were caused by progressive prostate cancer. Previously seen protective benefits of calcitriol with regard to serious adverse effects were not confirmed in the ASCENT-2 trial. There is no clear explanation as to why survival was lower in the treatment arm in ASCENT-1 vs. ASCENT-2 since regimens in calcitriol arm were identical.

Vitamin D analogs

Due to the potential of calcitriol to cause hypercalcemia and in order to improve on the anticancer effects of this compound, attempts have been made to develop more effective and less toxic analogs of vitamin D. In some cases preclinical studies suggested the usefulness of new analogs, and several clinical studies have been conducted using these agents.9092 Seocalcitol (EB1089) has been studied in phase I and II trials.

A phase I study was conducted in patients with advanced breast and colorectal cancers to assess the pharmacokinetic profile and toxicity of seocalcitol. While disease stabilization was noted in six patients for three months there were no other anti-tumor effects in that group of patients.90 The oral dose that led to hypercalciuria was considered dose-limiting toxicity and was subsequently used in the phase II study of the agent. These studies did not provide data that would support anti-tumor activity of seocalcitol on this dose and schedule in pancreatic or hepatocellular carcinoma patients.91,92

A phase I/II study of 19-nor-1alpha-25-dihydroxyvitamin D2 (paricalcitol) in advanced prostate cancer patients was conducted by Schwartz et al to assess the safety and efficacy of this vitamin D analog.93 The primary endpoint in this study was PSA response. Although the drug was generally well tolerated by patients at intravenous doses 3–15 μg/m2 three times weekly, no significant response was recorded.94

Most analogs tend to cause less hypercalcemia than calcitriol, but they also have a lower affinity for the vitamin D receptor, which in turn has been associated with reduction of anticancer properties.5

Conclusions

Several lines of evidence support the hypothesis that vitamin D and its metabolites might be beneficial to cancer chemoprevention and therapy. This relationship has been established based on in vitro and in vivo studies in tumor cells and animal models of different types of cancer, respectively. Many groups, including ours, have shown that high concentrations of biologically active vitamin D metabolites inhibit cancer cell proliferation and can induce differentation in some situations. The link between vitamin D and decreased incidence in several malignancies has also been strongly supported by epidemiologic studies.

Populations that are less likely to synthesize vitamin D based on their geographical location (due to lower levels of ultraviolet radiation) show higher risk of certain malignancies. This suggests that vitamin D provides protective effects against some malignancies. There are several serious limitations of testing calcitriol and other vitamin D metabolites in cancer prevention and treatment. It remains to be determined what the optimal biologic dose or concentration of calcitriol are. Second, most clinical studies did not use the optimal dose. Finally, due to drug formulation and other factors, clinical trials have not been designed adequately to assess the efficacy of calcitriol and analogs. Although results from preclinical studies strongly confirm anti-tumor properties of calcitriol, the lack of properly designed regimens has generated doubts about its clinical usefulness.

A number of clinical studies have been conducted with calcitriol or its analogs, but all are hampered by the above considerations. Many early studies of calcitriol and analogs employed daily, relatively low dose regimens. Little to no anti-tumor effects of limiting or at least worrisome hypercalcemia were seen. Our group and others have demonstrated that calcitriol exposure (AUC, Cmax) comparable to that achieved pre-clinically and which clearly results in anti-tumor effects is achieved with a high dose intermittent regimen. Results from clinical and preclinical studies show that high doses of calcitriol can be administered safely alone or in combination with other agents to elicit or enhance anti-tumor activity. Therefore, the rigorous testing of the anti-tumor effects of calcitriol should employ a high dose intermittent regimen of the dose approximately the MTD 100–120 μg/m2. No trials to date have used the MTD. Other dosing schedules, molecular endpoints, and interactions with other anticancer agents have not been thoroughly investigated.

Practice points.

  • Vitamin D deficiency/insufficiency are associated with increased cancer risk and poor outcome

  • Prospective trials to evaluate the effect of correcting deficiency/insufficiency are needed

  • High dose calcitriol is safe if used on an intermittent schedule

Research agenda.

  • Develop appropriate formulations of calcitriol for clinical use

  • Evaluate this formulation in a rigorous clinical development plan

    • Which cancer(s)

    • What clinical settings

    • With which chemotherapy regimens

  • Evaluate other analogs and determine the biologically optimal dose and schedule

Acknowledgments

AWR was supported by awards: R25CA113951-01 from the National Cancer Institute and W81WH11-1-0308 from the US Department of Defense.

CSJ was supported by awards: 5R01 CA085142-10, 5R01 CA067267-17, 5R01 CA095045-08 from the National Cancer Institute. DLT was supported by award: 5P30 CA016056-34 from the National Cancer Institute.

Footnotes

Conflict of interest statement

The authors have declared that no conflict of interest exists.

References

  • 1.Giovannucci E. Epidemiology of vitamin D and colorectal cancer: casual or causal link? The Journal of steroid biochemistry and molecular biology. 2010 Jul;121(1–2):349–354. doi: 10.1016/j.jsbmb.2010.03.085. [DOI] [PubMed] [Google Scholar]
  • 2.Giovannucci E. The epidemiology of vitamin D and cancer incidence and mortality: a review (United States) Cancer Causes and Control. 2005 Mar;16(2):83–95. doi: 10.1007/s10552-004-1661-4. [DOI] [PubMed] [Google Scholar]
  • 3.Purdue MP, Freedman DM, Gapstur SM, et al. Circulating 25-hydroxyvitamin D and risk of non-hodgkin lymphoma: cohort Consortium vitamin D Pooling Project of rarer cancers. American Journal of Epidemiology. 2010 Jul 1;172(1):58–69. doi: 10.1093/aje/kwq117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wei MY, Giovannucci EL. Vitamin D and multiple health outcomes in the Harvard cohorts. Molecular Nutrition & Food Research. 2010 Aug;54(8):1114–1126. doi: 10.1002/mnfr.200900574. [DOI] [PubMed] [Google Scholar]
  • *5.Deeb KK, Trump DL, Johnson CS. Vitamin D signalling pathways in cancer: potential for anticancer therapeutics. Nature Reviews Cancer. 2007 Sep;7(9):684–700. doi: 10.1038/nrc2196. [DOI] [PubMed] [Google Scholar]
  • 6.Brown AJ, Dusso A, Slatopolsky E. Vitamin D. American Journal of Physiology. 1999 Aug;277(2 Pt 2):F157–F175. doi: 10.1152/ajprenal.1999.277.2.F157. [DOI] [PubMed] [Google Scholar]
  • 7.Ahn J, Peters U, Albanes D, et al. Serum vitamin D concentration and prostate cancer risk: a nested case-control study. Journal of the National Cancer Institute. 2008 Jun 4;100(11):796–804. doi: 10.1093/jnci/djn152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Banach-Petrosky W, Ouyang X, Gao H, et al. Vitamin D inhibits the formation of prostatic intraepithelial neoplasia in Nkx3. 1; Pten mutant mice. Clinical Cancer Research. 2006 Oct 1;12(19):5895–5901. doi: 10.1158/1078-0432.CCR-06-1039. [DOI] [PubMed] [Google Scholar]
  • 9.Chung I, Han G, Seshadri M, et al. Role of vitamin D receptor in the antiproliferative effects of calcitriol in tumor-derived endothelial cells and tumor angiogenesis in vivo. Cancer Research. 2009 Feb 1;69(3):967–975. doi: 10.1158/0008-5472.CAN-08-2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Colston KW, James SY, Ofori-Kuragu EA, et al. Vitamin D receptors and anti-proliferative effects of vitamin D derivatives in human pancreatic carcinoma cells in vivo and in vitro. British Journal of Cancer. 1997;76(8):1017–1020. doi: 10.1038/bjc.1997.501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Getzenberg RH, Light BW, Lapco PE, et al. Vitamin D inhibition of prostate adenocarcinoma growth and metastasis in the Dunning rat prostate model system. Urology. 1997 Dec;50(6):999–1006. doi: 10.1016/S0090-4295(97)00408-1. [DOI] [PubMed] [Google Scholar]
  • 12.Jiang F, Bao J, Li P, et al. Induction of ovarian cancer cell apoptosis by 1,25-dihydroxyvitamin D3 through the down-regulation of telomerase. The Journal of Biological Chemistry. 2004 Dec 17;279(51):53213–53221. doi: 10.1074/jbc.M410395200. [DOI] [PubMed] [Google Scholar]
  • 13.Krishnan AV, Peehl DM, Feldman D. Inhibition of prostate cancer growth by vitamin D: regulation of target gene expression. Journal of Cellular Biochemistry. 2003 Feb 1;88(2):363–371. doi: 10.1002/jcb.10334. [DOI] [PubMed] [Google Scholar]
  • 14.Ma Y, Yu WD, Trump DL, Johnson CS. 1,25D3 enhances antitumor activity of gemcitabine and cisplatin in human bladder cancer models. Cancer. 2010 Jul 1;116(13):3294–3303. doi: 10.1002/cncr.25059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Nakagawa K, Kawaura A, Kato S, et al. 1 alpha,25-Dihydroxyvitamin D(3) is a preventive factor in the metastasis of lung cancer. Carcinogenesis. 2005 Feb;26(2):429–440. doi: 10.1093/carcin/bgh332. [DOI] [PubMed] [Google Scholar]
  • 16.Palmer HG, Sanchez-Carbayo M, Ordonez-Moran P, et al. Genetic signatures of differentiation induced by 1alpha, 25-dihydroxyvitamin D3 in human colon cancer cells. Cancer Research. 2003 Nov 15;63(22):7799–7806. [PubMed] [Google Scholar]
  • 17.Zhang X, Jiang F, Li P, et al. Growth suppression of ovarian cancer xenografts in nude mice by vitamin D analogue EB1089. Clinical Cancer Research. 2005 Jan 1;11(1):323–328. [PubMed] [Google Scholar]
  • 18.Holick MF. Vitamin D: a D-Lightful Solution for health. Journal of Investigative Medicine. 2011 Mar 16; doi: 10.231/JIM.0b013e318214ea2d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Holick MF. Vitamin D deficiency in 2010: health benefits of vitamin D and sunlight: a D-bate. Nature Reviews Endocrinology. 2011 Feb;7(2):73–75. doi: 10.1038/nrendo.2010.234. [DOI] [PubMed] [Google Scholar]
  • 20.Holick MF. Vitamin D: evolutionary, physiological and health perspectives. Current Drug Targets. 2011 Jan;12(1):4–18. doi: 10.2174/138945011793591635. [DOI] [PubMed] [Google Scholar]
  • *21.Dietary reference intakes for calcium and vitamin D. Institute of Medicine consensus report; Nov, 2010. [Google Scholar]
  • 22.Ross AC. The 2011 report on dietary reference intakes for calcium and vitamin D. Public Health Nutrition. 2011 May;14(5):938–939. doi: 10.1017/S1368980011000565. [DOI] [PubMed] [Google Scholar]
  • 23.Ross AC, Manson JE, Abrams SA, et al. The 2011 dietary reference intakes for calcium and vitamin d: what dietetics practitioners need to know. Journal of the American Dietetic Association. 2011 Apr;111(4):524–527. doi: 10.1016/j.jada.2011.01.004. [DOI] [PubMed] [Google Scholar]
  • 24.Ross AC, Manson JE, Abrams SA, et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. The Journal of Clinical Endocrinology & Metabolism. 2011 Jan;96(1):53–58. doi: 10.1210/jc.2010-2704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Heaney RP, Holick MF. Why the IOM recommendations for vitamin D are deficient. Journal of Bone and Mineral Research. 2011 Jan 4; doi: 10.1002/jbmr.328. [DOI] [PubMed] [Google Scholar]
  • 26.Arabi A, El Rassi R, El-Hajj Fuleihan G. Hypovitaminosis D in developing countries-prevalence, risk factors and outcomes. Nature Reviews Endocrinology. 2010 Oct;6(10):550–561. doi: 10.1038/nrendo.2010.146. [DOI] [PubMed] [Google Scholar]
  • *27.Garland CF, Garland FC. Do sunlight and vitamin D reduce the likelihood of colon cancer? International Journal of Epidemiology. 1980 Sep;9(3):227–231. doi: 10.1093/ije/9.3.227. [DOI] [PubMed] [Google Scholar]
  • 28.Ahonen MH, Tenkanen L, Teppo L, et al. Prostate cancer risk and prediagnostic serum 25-hydroxyvitamin D levels (Finland) Cancer Causes and Control. 2000 Oct;11(9):847–852. doi: 10.1023/a:1008923802001. [DOI] [PubMed] [Google Scholar]
  • 29.Bertone-Johnson ER, Chen WY, Holick MF, et al. Plasma 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D and risk of breast cancer. Cancer Epidemiology, Biomarkers & Prevention. 2005 Aug;14(8):1991–1997. doi: 10.1158/1055-9965.EPI-04-0722. [DOI] [PubMed] [Google Scholar]
  • 30.Cui Y, Rohan TE. Vitamin D, calcium, and breast cancer risk: a review. Cancer Epidemiology, Biomarkers & Prevention. 2006 Aug;15(8):1427–1437. doi: 10.1158/1055-9965.EPI-06-0075. [DOI] [PubMed] [Google Scholar]
  • 31.Garland CF, Comstock GW, Garland FC, et al. Serum 25-hydroxyvitamin D and colon cancer: eight-year prospective study. Lancet. 1989 Nov 18;2(8673):1176–1178. doi: 10.1016/s0140-6736(89)91789-3. [DOI] [PubMed] [Google Scholar]
  • 32.Giovannucci E, Liu Y, Rimm EB, et al. Prospective study of predictors of vitamin D status and cancer incidence and mortality in men. Journal of the National Cancer Institute. 2006 Apr 5;98(7):451–459. doi: 10.1093/jnci/djj101. [DOI] [PubMed] [Google Scholar]
  • 33.McCullough ML, Weinstein SJ, Freedman DM, et al. Correlates of circulating 25-hydroxyvitamin D: cohort Consortium vitamin D Pooling Project of rarer cancers. American Journal of Epidemiology. 2010 Jul 1;172(1):21–35. doi: 10.1093/aje/kwq113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zeleniuch-Jacquotte A, Gallicchio L, Hartmuller V, et al. Circulating 25-hydroxyvitamin D and risk of endometrial cancer: cohort Consortium vitamin D Pooling Project of rarer cancers. American Journal of Epidemiology. 2010 Jul 1;172(1):36–46. doi: 10.1093/aje/kwq114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gallicchio L, Helzlsouer KJ, Chow WH, et al. Circulating 25-hydroxyvitamin D and the risk of rarer cancers: design and methods of the cohort Consortium vitamin D Pooling Project of rarer cancers. American Journal of Epidemiology. 2010 Jul 1;172(1):10–20. doi: 10.1093/aje/kwq116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gallicchio L, Moore LE, Stevens VL, et al. Circulating 25-hydroxyvitamin D and risk of kidney cancer: cohort consortium vitamin D Pooling Project of rarer cancers. American Journal of Epidemiology. 2010 Jul 1;172(1):47–57. doi: 10.1093/aje/kwq115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zheng W, Danforth KN, Tworoger SS, et al. Circulating 25-hydroxyvitamin D and risk of epithelial ovarian cancer: cohort consortium vitamin D pooling project of rarer cancers. American Journal of Epidemiology. 2010 Jul 1;172(1):70–80. doi: 10.1093/aje/kwq118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Stolzenberg-Solomon RZ, Jacobs EJ, Arslan AA, et al. Circulating 25-hydroxyvitamin D and risk of pancreatic cancer: cohort consortium vitamin D pooling project of rarer cancers. American Journal of Epidemiology. 2010 Jul 1;172(1):81–93. doi: 10.1093/aje/kwq120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Mondul AM, Weinstein SJ, Mannisto S, et al. Serum vitamin D and risk of bladder cancer. Cancer Research. 2010 Nov 15;70(22):9218–9223. doi: 10.1158/0008-5472.CAN-10-0985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Konety BR, Lavelle JP, Pirtskalaishvili G, et al. Effects of vitamin D (calcitriol) on transitional cell carcinoma of the bladder in vitro and in vivo. Journal of Urology. 2001 Jan;165(1):253–258. doi: 10.1097/00005392-200101000-00074. [DOI] [PubMed] [Google Scholar]
  • 41.Higashi T, Homma S, Iwata H, et al. Characterization of urinary metabolites of vitamin D(3) in man under physiological conditions using liquid chromatography-tandem mass spectrometry. Journal of Pharmaceutical and Biomedical Analysis. 2002 Jul 31;29(5):947–955. doi: 10.1016/s0731-7085(02)00135-8. [DOI] [PubMed] [Google Scholar]
  • 42.Thierry-Palmer M, Henderson VM, Hammali RE, et al. Black and white female adolescents lose vitamin D metabolites into urine. The American Journal of the Medical Sciences. 2008 Apr;335(4):278–283. doi: 10.1097/MAJ.0b013e31815768db. [DOI] [PubMed] [Google Scholar]
  • 43.Tanaka H, Abe E, Miyaura C, et al. 1 alpha,25-Dihydroxycholecalciferol and a human myeloid leukaemia cell line (HL-60) Biochemical Journal. 1982 Jun 15;204(3):713–719. doi: 10.1042/bj2040713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Miyaura C, Abe E, Kuribayashi T, et al. 1 alpha,25-Dihydroxyvitamin D3 induces differentiation of human myeloid leukemia cells. Biochemical and Biophysical Research Communications. 1981 Oct 15;102(3):937–943. doi: 10.1016/0006-291x(81)91628-4. [DOI] [PubMed] [Google Scholar]
  • 45.Abe E, Miyaura C, Sakagami H, et al. Differentiation of mouse myeloid leukemia cells induced by 1 alpha, 25-dihydroxyvitamin D3. Proceedings of the National Academy of Sciences. 1981 Aug;78(8):4990–4994. doi: 10.1073/pnas.78.8.4990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Krishnan AV, Trump DL, Johnson CS, et al. The role of vitamin D in cancer prevention and treatment. Endocrinology Metabolism Clinics of North America. 2010 Jun;39(2):401–418. doi: 10.1016/j.ecl.2010.02.011. table of contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Trump DL, Deeb KK, Johnson CS. Vitamin D: considerations in the continued development as an agent for cancer prevention and therapy. The Cancer Journal. 2010 Jan-Feb;16(1):1–9. doi: 10.1097/PPO.0b013e3181c51ee6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Beer TM, Lemmon D, Lowe BA, et al. High-dose weekly oral calcitriol in patients with a rising PSA after prostatectomy or radiation for prostate carcinoma. Cancer. 2003 Mar 1;97(5):1217–1224. doi: 10.1002/cncr.11179. [DOI] [PubMed] [Google Scholar]
  • 49.Gross C, Stamey T, Hancock S, et al. Treatment of early recurrent prostate cancer with 1,25-dihydroxyvitamin D3 (calcitriol) Journal of Urology. 1998 Jun;159(6):2035–2039. doi: 10.1016/S0022-5347(01)63236-1. discussion 9–40. [DOI] [PubMed] [Google Scholar]
  • *50.Trump DL, Potter DM, Muindi J, et al. Phase II trial of high-dose, intermittent calcitriol (1,25 dihydroxyvitamin D3) and dexamethasone in androgen-independent prostate cancer. Cancer. 2006 May 15;106(10):2136–2142. doi: 10.1002/cncr.21890. [DOI] [PubMed] [Google Scholar]
  • 51.Beer TM, Myrthue A. Calcitriol in cancer treatment: from the lab to the clinic. Molecular Cancer Therapeutics. 2004 Mar;3(3):373–381. [PubMed] [Google Scholar]
  • *52.Beer TM, Javle MM, Ryan CW, et al. Phase I study of weekly DN-101, a new formulation of calcitriol, in patients with cancer. Cancer Chemotherapy and Pharmacology. 2007 Apr;59(5):581–587. doi: 10.1007/s00280-006-0299-1. [DOI] [PubMed] [Google Scholar]
  • 53.Chadha MK, Tian L, Mashtare T, et al. Phase 2 trial of weekly intravenous 1,25 dihydroxy cholecalciferol (calcitriol) in combination with dexamethasone for castration-resistant prostate cancer. Cancer. 2010 May 1;116(9):2132–2139. doi: 10.1002/cncr.24973. [DOI] [PubMed] [Google Scholar]
  • *54.Fakih MG, Trump DL, Muindi JR, et al. A phase I pharmacokinetic and pharmacodynamic study of intravenous calcitriol in combination with oral gefitinib in patients with advanced solid tumors. Clinical Cancer Research. 2007 Feb 15;13(4):1216–1223. doi: 10.1158/1078-0432.CCR-06-1165. [DOI] [PubMed] [Google Scholar]
  • 55.Muindi JR, Johnson CS, Trump DL, et al. A phase I and pharmacokinetics study of intravenous calcitriol in combination with oral dexamethasone and gefitinib in patients with advanced solid tumors. Cancer Chemotherapy and Pharmacology. 2009 Dec;65(1):33–40. doi: 10.1007/s00280-009-1000-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Muindi JR, Peng Y, Potter DM, et al. Pharmacokinetics of high-dose oral calcitriol: results from a phase 1 trial of calcitriol and paclitaxel. Clinical Pharmacology & Therapeutics. 2002 Dec;72(6):648–659. doi: 10.1067/mcp.2002.129305. [DOI] [PubMed] [Google Scholar]
  • 57.Osborn JL, Schwartz GG, Smith DC, et al. Phase II trial of oral 1,25-dihydroxyvitamin D (calcitriol) in hormone refractory prostate cancer. Urologic Oncology. 1995 Sep-Oct;1(5):195–198. doi: 10.1016/1078-1439(95)00061-5. [DOI] [PubMed] [Google Scholar]
  • 58.Wilding G, Soulie P, Trump D, et al. Results from a pilot Phase I trial of gefitinib combined with docetaxel and estramustine in patients with hormone-refractory prostate cancer. Cancer. 2006 May 1;106(9):1917–1924. doi: 10.1002/cncr.21831. [DOI] [PubMed] [Google Scholar]
  • *59.Beer TM, Ryan CW, Venner PM, et al. Double-blinded randomized study of high-dose calcitriol plus docetaxel compared with placebo plus docetaxel in androgen-independent prostate cancer: a report from the ASCENT Investigators. Journal of Clinical Oncology. 2007 Feb 20;25(6):669–674. doi: 10.1200/JCO.2006.06.8197. [DOI] [PubMed] [Google Scholar]
  • *60.Scher HI, Jia X, Chi K, et al. Randomized, Open-Label phase III trial of docetaxel plus high-dose calcitriol versus docetaxel plus prednisone for patients with castration-resistant prostate cancer. Journal of Clinical Oncology. 2011 Apr 11; doi: 10.1200/JCO.2010.32.8815. [DOI] [PubMed] [Google Scholar]
  • 61.Muindi JR, Potter DM, Peng Y, et al. Pharmacokinetics of liquid calcitriol formulation in advanced solid tumor patients: comparison with caplet formulation. Cancer Chemotherapy and Pharmacology. 2005 Nov;56(5):492–496. doi: 10.1007/s00280-005-1015-2. [DOI] [PubMed] [Google Scholar]
  • 62.Beer TM, Munar M, Henner WD. A Phase I trial of pulse calcitriol in patients with refractory malignancies: pulse dosing permits substantial dose escalation. Cancer. 2001 Jun 15;91(12):2431–2439. [PubMed] [Google Scholar]
  • 63.Hellstrom E, Robert KH, Samuelsson J, et al. Treatment of myelodysplastic syndromes with retinoic acid and 1 alpha-hydroxy-vitamin D3 in combination with low-dose ara-C is not superior to ara-C alone. Results from a randomized study. The Scandinavian Myelodysplasia Group (SMG) European Journal of Haematology. 1990 Nov;45(5):255–261. doi: 10.1111/j.1600-0609.1990.tb00470.x. [DOI] [PubMed] [Google Scholar]
  • 64.Petrini M, Caracciolo F, Corini M, et al. Low-dose ARA-C and 1(OH) D3 administration in acute non lymphoid leukemia: pilot study. Haematologica. 1991 May-Jun;76(3):200–203. [PubMed] [Google Scholar]
  • 65.Ferrero D, Bruno B, Pregno P, et al. Combined differentiating therapy for myelodysplastic syndromes: a phase II study. Leukemia Research. 1996 Oct;20(10):867–876. doi: 10.1016/0145-2126(95)00156-5. [DOI] [PubMed] [Google Scholar]
  • 66.Slapak CA, Desforges JF, Fogaren T, et al. Treatment of acute myeloid leukemia in the elderly with low-dose cytarabine, hydroxyurea, and calcitriol. American Journal of Hematology. 1992 Nov;41(3):178–183. doi: 10.1002/ajh.2830410307. [DOI] [PubMed] [Google Scholar]
  • 67.Masuda S, Jones G. Vitamin D analogs–drug design based on proteins involved in vitamin D signal transduction. Current Drug Targets - Immune Endocrine & Metabolic Disorders. 2003 Mar;3(1):43–66. [PubMed] [Google Scholar]
  • 68.Hansen CM, Hamberg KJ, Binderup E, et al. Seocalcitol (EB 1089): a vitamin D analogue of anti-cancer potential. Background, design, synthesis, pre-clinical and clinical evaluation. Current Pharmaceutical Design. 2000 May;6(7):803–828. doi: 10.2174/1381612003400371. [DOI] [PubMed] [Google Scholar]
  • 69.Bareis P, Bises G, Bischof MG, et al. 25-hydroxy-vitamin d metabolism in human colon cancer cells during tumor progression. Biochemical and Biophysical Research Communications. 2001 Jul 27;285(4):1012–1017. doi: 10.1006/bbrc.2001.5289. [DOI] [PubMed] [Google Scholar]
  • 70.Chen TC, Wang L, Whitlatch LW, et al. Prostatic 25-hydroxyvitamin D-1alpha-hydroxylase and its implication in prostate cancer. Journal of Cellular Biochemistry. 2003 Feb 1;88(2):315–322. doi: 10.1002/jcb.10342. [DOI] [PubMed] [Google Scholar]
  • 71.Nomura AM, Stemmermann GN, Lee J, et al. Serum vitamin D metabolite levels and the subsequent development of prostate cancer (Hawaii, United States) Cancer Causes and Control. 1998 Aug;9(4):425–432. doi: 10.1023/a:1008875819232. [DOI] [PubMed] [Google Scholar]
  • 72.Segersten U, Correa P, Hewison M, et al. 25-hydroxyvitamin D(3)-1alpha-hydroxylase expression in normal and pathological parathyroid glands. The Journal of Clinical Endocrinology & Metabolism. 2002 Jun;87(6):2967–2972. doi: 10.1210/jcem.87.6.8604. [DOI] [PubMed] [Google Scholar]
  • 73.Ding EL, Mehta S, Fawzi WW, et al. Interaction of estrogen therapy with calcium and vitamin D supplementation on colorectal cancer risk: reanalysis of Women’s Health Initiative randomized trial. International Journal of Cancer. 2008 Apr 15;122(8):1690–1694. doi: 10.1002/ijc.23311. [DOI] [PubMed] [Google Scholar]
  • 74.Woo TC, Choo R, Jamieson M, et al. Pilot study: potential role of vitamin D (Cholecalciferol) in patients with PSA relapse after definitive therapy. Nutrition and Cancer. 2005;51(1):32–36. doi: 10.1207/s15327914nc5101_5. [DOI] [PubMed] [Google Scholar]
  • 75.Amir E, Simmons CE, Freedman OC, et al. A phase 2 trial exploring the effects of high-dose (10,000 IU/day) vitamin D(3) in breast cancer patients with bone metastases. Cancer. 2010 Jan 15;116(2):284–291. doi: 10.1002/cncr.24749. [DOI] [PubMed] [Google Scholar]
  • 76.Smith DC, Johnson CS, Freeman CC, et al. A Phase I trial of calcitriol (1,25-dihydroxycholecalciferol) in patients with advanced malignancy. Clinical Cancer Research. 1999 Jun;5(6):1339–1345. [PubMed] [Google Scholar]
  • 77.Beer TM. Development of weekly high-dose calcitriol based therapy for prostate cancer. Urologic Oncology. 2003 Sep-Oct;21(5):399–405. doi: 10.1016/s1078-1439(03)00170-4. [DOI] [PubMed] [Google Scholar]
  • *78.Beer TM, Javle M, Lam GN, et al. Pharmacokinetics and tolerability of a single dose of DN-101, a new formulation of calcitriol, in patients with cancer. Clinical Cancer Research. 2005 Nov 1;11(21):7794–7799. doi: 10.1158/1078-0432.CCR-05-0552. [DOI] [PubMed] [Google Scholar]
  • 79.Rassnick KM, Muindi JR, Johnson CS, et al. In vitro and in vivo evaluation of combined calcitriol and cisplatin in dogs with spontaneously occurring tumors. Cancer Chemotherapy and Pharmacology. 2008 Oct;62(5):881–891. doi: 10.1007/s00280-008-0678-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Abe-Hashimoto J, Kikuchi T, Matsumoto T, et al. Antitumor effect of 22-oxa-calcitriol, a noncalcemic analogue of calcitriol, in athymic mice implanted with human breast carcinoma and its synergism with tamoxifen. Cancer Research. 1993 Jun 1;53(11):2534–2537. [PubMed] [Google Scholar]
  • 81.Sternberg CN, Donat SM, Bellmunt J, et al. Chemotherapy for bladder cancer: treatment guidelines for neoadjuvant chemotherapy, bladder preservation, adjuvant chemotherapy, and metastatic cancer. Urology. 2007 Jan;69(1):62–79. doi: 10.1016/j.urology.2006.10.041. [DOI] [PubMed] [Google Scholar]
  • 82.Rashid SF, Moore JS, Walker E, et al. Synergistic growth inhibition of prostate cancer cells by 1 alpha,25 Dihydroxyvitamin D(3) and its 19-nor-hexafluoride analogs in combination with either sodium butyrate or trichostatin A. Oncogene. 2001 Apr 5;20(15):1860–1872. doi: 10.1038/sj.onc.1204269. [DOI] [PubMed] [Google Scholar]
  • 83.Swami S, Krishnan AV, Peehl DM, et al. Genistein potentiates the growth inhibitory effects of 1,25-dihydroxyvitamin D3 in DU145 human prostate cancer cells: role of the direct inhibition of CYP24 enzyme activity. Molecular and Cellular Endocrinology. 2005 Sep 28;241(1–2):49–61. doi: 10.1016/j.mce.2005.05.001. [DOI] [PubMed] [Google Scholar]
  • 84.Flaig TW, Barqawi A, Miller G, et al. A phase II trial of dexamethasone, vitamin D, and carboplatin in patients with hormone-refractory prostate cancer. Cancer. 2006 Jul 15;107(2):266–274. doi: 10.1002/cncr.21982. [DOI] [PubMed] [Google Scholar]
  • 85.Beer TM, Eilers KM, Garzotto M, et al. Weekly high-dose calcitriol and docetaxel in metastatic androgen-independent prostate cancer. Journal of Clinical Oncology. 2003 Jan 1;21(1):123–128. doi: 10.1200/jco.2003.05.117. [DOI] [PubMed] [Google Scholar]
  • *86.Beer TM, Hough KM, Garzotto M, et al. Weekly high-dose calcitriol and docetaxel in advanced prostate cancer. Seminars in Oncology. 2001 Aug;28(4 Suppl 15):49–55. doi: 10.1016/s0093-7754(01)90155-1. [DOI] [PubMed] [Google Scholar]
  • 87.Light BW, Yu WD, McElwain MC, et al. Potentiation of cisplatin antitumor activity using a vitamin D analogue in a murine squamous cell carcinoma model system. Cancer Research. 1997 Sep 1;57(17):3759–3764. [PubMed] [Google Scholar]
  • 88.Hershberger PA, Yu WD, Modzelewski RA, et al. Calcitriol (1,25-dihydroxycholecalciferol) enhances paclitaxel antitumor activity in vitro and in vivo and accelerates paclitaxel-induced apoptosis. Clinical Cancer Research. 2001 Apr;7(4):1043–1051. [PubMed] [Google Scholar]
  • 89.Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. The New England Journal of Medicine. 2004 Oct 7;351(15):1502–1512. doi: 10.1056/NEJMoa040720. [DOI] [PubMed] [Google Scholar]
  • 90.Gulliford T, English J, Colston KW, et al. A phase I study of the vitamin D analogue EB 1089 in patients with advanced breast and colorectal cancer. British Journal of Cancer. 1998 Jul;78(1):6–13. doi: 10.1038/bjc.1998.434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Evans TR, Colston KW, Lofts FJ, et al. A phase II trial of the vitamin D analogue Seocalcitol (EB1089) in patients with inoperable pancreatic cancer. British Journal of Cancer. 2002 Mar 4;86(5):680–685. doi: 10.1038/sj.bjc.6600162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Dalhoff K, Dancey J, Astrup L, et al. A phase II study of the vitamin D analogue Seocalcitol in patients with inoperable hepatocellular carcinoma. British Journal of Cancer. 2003 Jul 21;89(2):252–257. doi: 10.1038/sj.bjc.6601104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Schwartz GG, Eads D, Naczki C, et al. 19-nor-1 alpha,25-dihydroxyvitamin D2 (paricalcitol) inhibits the proliferation of human pancreatic cancer cells in vitro and in vivo. Cancer Biology and Therapy. 2008 Mar;7(3):430–436. doi: 10.4161/cbt.7.3.5418. [DOI] [PubMed] [Google Scholar]
  • 94.Schwartz GG, Hall MC, Stindt D, et al. Phase I/II study of 19-nor-1alpha-25-dihydroxyvitamin D2 (paricalcitol) in advanced, androgen-insensitive prostate cancer. Clinical Cancer Research. 2005 Dec 15;11(24 Pt 1):8680–8685. doi: 10.1158/1078-0432.CCR-05-1237. [DOI] [PubMed] [Google Scholar]

RESOURCES