U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Some Antiviral and Antineoplastic Drugs, and Other Pharmaceutical Agents. Lyon (FR): International Agency for Research on Cancer; 2000. (IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, No. 76.)

Cover of Some Antiviral and Antineoplastic Drugs, and Other Pharmaceutical Agents

Some Antiviral and Antineoplastic Drugs, and Other Pharmaceutical Agents.

Show details

Other pharmaceutical agents

Hydroxyurea

1. Exposure Data

1.1. Chemical and physical data

1.1.1. Nomenclature
  • Chem. Abstr. Serv. Reg. No.: 127-07-1
  • Chem. Abstr. Name: Hydroxyurea
  • IUPAC Systematic Name: Hydroxyurea
  • Synonyms: N-(Aminocarbonyl)hydroxylamine; carbamohydroxamic acid; carbamohydroximic acid; carbamoyl oxime; HU; hydroxycarbamide; hydroxycarbamine; hydroxylurea
1.1.2. Structural and molecular formulae and relative molecular mass
Image 347a
1.1.3. Chemical and physical properties of the pure substance
1.1.4. Technical products and impurities

Hydroxyurea is available as a 200-, 300-, 400- or 500-mg capsule; the capsule may also contain calcium citrate, citric acid, colourants (D&C Red No. 28; D&C Red No. 33; D&C Yellow No. 10; FD&C Blue No.1; FD&C Green No. 3; FD&C Red No. 40), disodium citrate, erythrosine, gelatin, indigocarmine, iron oxide, lactose, magnesium stearate, sodium lauryl sulfate, sodium monohydrogen phosphate, tartrazine and titanium dioxide (American Hospital Formulary Service, 1997; British Medical Association/Royal Pharmaceutical Society of Great Britain, 1998; Editions du Vidal, 1998; Rote Liste Sekretariat, 1998; Thomas, 1998; US Pharmacopeial Convention, 1998; Medical Economics Data Production, 1999).

Trade names for hydroxyurea include Biosupressin, Droxia, Droxiurea, Hidroks, Hidroxiurea Asofarma, Hidroxiurea Filaxis, Hidroxiurea Martian, Hydrea, Hydrea capsules, Hydreia, Hydroxycarbamid, Hydroxycarbamide capsules BP 1998, Hydroxycarbamide capsules USP 23, Hydroxyurea, Litalir, Onco-Carbide, Oxyurea and Syrea (CIS Information Services, 1998; Royal Pharmaceutical Society of Great Britain, 1999; Swiss Pharmaceutical Society, 1999).

1.1.5. Analysis

Several international pharmacopoeias specify infrared absorption spectrophotometry with comparison to standards as the method for identifying hydroxyurea; titration with sodium thiosulfate is used to assay its purity. Similar methods are used for identifying and assaying hydroxyurea in pharmaceutical preparations (British Pharmacopoeial Commission, 1993; US Pharmacopeial Convention, 1994).

1.2. Production

Hydroxyurea has been prepared by the reaction of calcium cyanate with hydroxylamine nitrate in absolute ethanol and by the reaction of potassium cyanate and hydroxylamine hydrochloride in aqueous solution. Hydroxyurea has also been prepared by converting a quaternary ammonium anion exchange resin from the chloride form to the cyanate form with sodium cyanate and reacting the resin in the cyanate form with hydroxylamine hydrochloride (Graham, 1955).

Information available in 1999 indicated that hydroxyurea was manufactured and/or formulated in 25 countries (CIS Information Services, 1998; Royal Pharmaceutical Society of Great Britain; 1999; Swiss Pharmaceutical Society, 1999).

1.3. Use

Hydroxyurea is a chemically simple antimetabolite, which is cytostatic by inhibiting ribonucleotide reductase, an enzyme important in creating deoxynucleosides for DNA replication in growing cells (Gao et al., 1998). Hydroxyurea was initially synthesized over 120 years ago, but its potential biological significance was not recognized until 1928. In the late 1950s, the drug was evaluated in a large number of experimental murine tumour systems and shown to be active against a broad spectrum of tumours. Phase I trials with hydroxyurea began in 1960 and by the late 1960s it was in clinical use (Donehower, 1992).

Hydroxyurea has been used, or investigated for use, in the treatment of a number of diseases.

(a) Sickle-cell haemoglobinopathy

Hydroxyurea is widely used to treat severe sickle-cell disease (Charache et al., 1996; Ferster et al., 1996; de Montalembert et al., 1997) and beta-thalessaemia–sickle-cell disease (Voskaridou et al., 1995). It was shown to induce fetal haemoglobin synthesis (Fibach et al., 1993; Maier-Redelsperger et al., 1998), and preliminary reports demonstrated its benefit in beta-thalessaemia–sickle-cell haemoglobinopathy (Loukopoulos et al., 1998). The efficacy of hydroxyurea in sickle-cell disease is well validated, but its use appears to be limited to patients with frequent crises and hospitalizations. The doses used in patients with sickle-cell anaemia are 25 mg/kg bw per day in children (Ferster et al., 1996) and up to 35 mg/kg bw per day in adults (Charache et al., 1996).

(b) Myeloproliferative syndromes

Hydroxyurea is also used as a cytostatic agent in myelodyplastic or myeloproliferative diseases, including chronic myeloid leukaemia (Donehower, 1992; Fitzgerald & McCann, 1993; Guilhot et al., 1993), polycythemia vera (Najean & Rain, 1997a,b), myelodysplastic syndrome (Nair et al., 1993) and essential thrombocythaemia and corticosteroid-resistant hypereosinophilia (Donehower, 1992). The risks and benefits of hydroxyurea in haematological disease are debated and have been reviewed (Donehower, 1992). It is used most commonly in chronic myeloid leukaemia to prevent or delay the onset of blast crises, and complete responses have been seen occasionally (Tanaka et al., 1997).

(c) With didanosine in the treatment of HIV/AIDS

Hydroxyurea is given as an adjunct with didanosine (see monograph, this volume) in treatment of human immunodeficiency virus (HIV) infection. Several case series and randomized trials have shown dramatic results with the combination (Biron et al., 1996; Montaner et al., 1997; Foli et al., 1998), although not without exception (Simonelli et al., 1997). Trials in which didanosine and hydroxyurea were given in combination with other agents (Lisziewicz et al., 1998; Rutschmann et al., 1998) led to large-scale controlled trials which are currently under way. Intriguing reports of prolonged periods without rebound viraemia after didanosine and hydroxyurea therapy, especially in patients who began treatment shortly after HIV infection, have led to intensive investigation (Vila et al., 1997; Lisziewicz et al., 1998). The doses of hydroxyurea used in combination with didanosine are 500–1000 mg/day (Montaner et al., 1997; Rutschmann et al., 1998).

Hydroxyurea itself has no antiviral effect (Lori et al., 1997a). Its mechanism of action with didanosine appears to be selective inhibition of ribonucleotide reductase, thus decreasing endogenous dATP concentrations, leading to increased generation of dATP from the pro-drug didanosine. Hydroxyurea thus improves the antiviral potency of didanosine (Lori et al., 1997b; De Boer et al., 1998; Johns & Gao, 1998). The effect is likely to be greater with dATP analogues such as didanosine than with other antiviral nucleoside analogues (Gandhi et al., 1998).

Increased cytokine levels (Navarra et al., 1995, 1996) and adrenal activity have been seen in hydroxyurea-treated patients (Navarra et al., 1990, 1998) but not in HIV-infected patients treated with hydroxyurea and didanosine.

(d) Psoriasis

Hydroxyurea can be administered over long periods to treat psoriasis (Moschella & Greenwald, 1973; Boyd & Neldner, 1991), although it is currently used relatively infrequently.

(e) Solid tumours

Hydroxyurea is used as a radiosensitizing agent in carcinoma of the cervix (Stehman, 1992; Stehman et al., 1997) and glioma (Levin & Prados, 1992). Other malignancies in which use of hydroxyurea as an adjunct has been studied (Wadler et al., 1996) include meningioma (Schrell et al., 1997), uterine leiomyosarcoma (Currie et al., 1996a) and uterine mixed mesodermal tumours (Currie et al., 1996b).

1.4. Occurrence

Hydroxyurea is not known to occur as a natural product. No data on occupational exposure were available to the Working Group.

1.5. Regulations and guidelines

Hydroxyurea is listed in the British, French and US pharmacopoeias (Royal Pharmaceutical Society of Great Britain, 1999; Swiss Pharmaceutical Society, 1999).

2. Studies of Cancer in Humans

The carcinogenic potential of hydroxyurea has been studied in patients with chronic myeloproliferative disorders, which include chronic myeloid leukaemia, ideopathic myelofibrosis, polycythaemia vera and essential thrombocythaemia. An assessment of the carcinogenicity of this agent is hampered by an inherent tendency of chronic myeloproliferative disorders to undergo spontaneous transformation to myelodysplastic syndrome or acute leukaemia. Thus, among 431 patients with polycythaemia vera who were randomized to one of three treatment arms, phlebotomy (n = 134), chlorambucil (n = 141) or radioactive phosphorus (n = 156), patients in the phlebotomy arm were found to have a cumulative risk for acute leukaemia after 11–18 years of follow-up of 1.5% on the basis of two observed cases. The rates for acute myeloid leukaemia in the US population indicate that about 0.13 cases would have been expected (Landaw, 1986).

Although hydroxyurea is used in the treatment of sickle-cell anaemia and of psoriasis, these conditions are not suspected to predispose to cancer.

2.1. Case reports

Several case reports have been published on the occurrence of multiple squamous-cell and basal-cell carcinomas of the skin in patients who received prolonged treatment with hydroxyurea for chronic myeloid leukaemia (Disdier et al., 1991; Stasi et al., 1992; Best & Petitt, 1998; De Simone et al., 1998), essential thrombocythaemia or polycythaemia vera (Callot-Mellot et al., 1996; Best & Petitt, 1998). The skin carcinomas were typically seen in sun-exposed areas and had been preceded by other degenerative cutaneous manifestations.

Reiffers et al. (1985), van den Anker-Lugtenburg and Sizoo (1990) and Furgerson et al. (1996) described the occurrence of acute leukaemia in patients who had been treated with hydroxyurea for long periods for essential thrombocythaemia and had not received other treatments.

2.2. Cohort studies

These studies are summarized in Table 1.

Table 1. Cohort studies of acute leukaemia (AL) and myelodysplastic syndrome (MDS) in patients treated with hydroxyurea.

Table 1

Cohort studies of acute leukaemia (AL) and myelodysplastic syndrome (MDS) in patients treated with hydroxyurea.

2.2.1. Polycythaemia vera (see also section 2.2.3)

Sharon et al. (1986) reported the results of a prospective study conducted in Israel of 36 patients with polycythaemia vera who were treated with hydroxyurea at a daily dose of 500–1500 mg for 1–5.6 years. Nineteen of the patients had previously been treated with other myelosuppressive drugs. During treatment, no cases of leukaemia or other malignant neoplasms were seen.

West (1987) studied the incidence of acute leukaemia in 100 patients in Kentucky, USA, who were treated for polycythaemia vera which had been diagnosed during 1963–83. They were also treated by phlebotomy. The mean daily dose of hydroxyurea was 720 mg, and the duration of therapy ranged from three months to 18 years (mean, 5.4 years). During this time, two (2%) cases of leukaemia were observed: one chronic neutrophilic leukaemia after nine months of treatment and one acute myeloid leukaemia after five years. The authors noted that the chronic neutrophilic leukaemia might have been present before the date of recruitment.

Of 118 patients in New York, USA, with polycythaemia vera, all had received supplementary phlebotomy and hydroxyurea at a daily dose of 30 mg/kg bw for one week, then 15 mg/kg bw and then modified downwards and upwards; 59 of the patients had had no prior myelosuppressive therapy (Donovan et al., 1984). Hydroxyurea was given for a mean of 4.7 years (range, one month to 7.5 years). Three out of 51 patients with no prior myelosuppressive therapy developed acute leukaemia after 1.7, 2.8 and 4.8 years of treatment (Kaplan et al., 1986). Two cases of leukaemia (1.5%) were observed in the most appropriate historical control group of 134 patients who had been treated exclusively with phlebotomy (Landaw, 1986), and the difference between the two groups was not statistically significant. [The Working Group noted the reduction in numbers of hydroxyurea-treated patients from 59 to 51, which was unexplained.] In a subsequent case report, Holcombe et al. (1991) described an additional case of chronic myelomonocytic leukaemia (which transforms to acute myeloid leukaemia) in the group of 51 hydroxyurea-treated patients with polycythaemia vera. This case was seen 19 years from the date of initial diagnosis. No data were given on the historical control group.

Nand et al. (1990) conducted a retrospective study of 48 patients in Chicago, Illinois, USA, with polycythaemia vera diagnosed during 1975–87 (seven cases were diagnosed previously) and treated over a period of 12 years. Of these, 18 had been treated with hydroxyurea at doses ranging from 500 mg every other day for nine months to 1000 mg daily for seven years. Four cases of acute leukaemia were seen among 10 of the patients who had received hydroxyurea in combination with other myelosuppressive treatment including radioactive phosphorus and one case among eight patients who had received hydroxyurea alone (relative risk = 3.8; p = 0.38). The cases developed at a mean of 3.9 years after the start of treatment. [The Working Group noted the small size of the study.]

Two published reports are available on the results of a clinical trial of the French Polycythaemia Vera Study Group (Najean et al., 1996; Najean & Rain, 1997a). The most recent report includes 292 patients with polycythaemia vera diagnosed after 1980 when the patients were aged 0–64 years. The patients were treated with either hydroxyurea (n = 150) at a daily dose of 25 mg/kg bw followed by a maintenance dose of 10–15 mg/kg bw, or pipobroman [1,4-bis(3-bromopropionyl)piperazine] (n = 142). The patients were followed for 1–17 years, during which time nine cases of acute myeloid leukaemia and four cases of myelodysplastic syndrome were observed. The precise treatment received in these 13 cases was not specified, but the authors stated that the actuarial risk for acute myeloid leukaemia or myelodysplastic syndrome was about 10% at the 13th year of follow-up, with no difference according to treatment. Four cases of non-melanoma skin cancer were seen in patients given hydroxyurea only and one in a patient given pipobroman only, while six cancers at sites other than the bone marrow and non-melanoma skin were seen in six patients given hydroxyurea and five given pipobroman. These frequencies of extracutaneous solid tumours were only slightly greater than those expected for this age group.

In a complementary trial covering the period 1979–96, Najean and Rain (1997b) recruited 461 patients with polycythaemia vera who were over 65 years of age and had not previously been treated with chemotherapeutic agents. Initially, the patients were treated with radioactive phosphorus (0.1 mCi/kg bw with a maximum of 7 mCi) administered intravenously until complete remission of the polycythaemia was obtained. They were then randomized to receive either maintenance treatment with low-dose hydroxyurea (5–10 mg/kg bw per day) (n = 219) or simple surveillance (n = 242). When the haematocrit of a patient in either treatment arm had increased to 50% and the erythrocyte volume was > 125% of the normal value during follow-up, intravenous administration of radioactive phosphorus was resumed. The median survival was 9.1 years in the group receiving maintenance therapy and 11.2 years in the surveyed group (p = 0.10). In a subset of 408 patients followed for more than two years (maximum follow-up time, 16 years), the mean annual dose of radioactive phosphorus was 0.009 mCi/kg bw in the group receiving hydroxyurea and 0.033 mCi/kg bw in the surveyed group [average for all study subjects, approximately 0.021 mCi/kg bw per year]. In the same subset of 408 patients, 41 haematological malignancies were observed, consisting of 15 cases of acute myeloid leukaemia, two of non-Hodgkin lymphoma, two of chronic lymphocytic leukaemia, two of multiple myeloma, three of chronic myelomonocytic leukaemia and 17 of myelodysplastic syndrome. The precise treatment schedule received in these 41 cases was not specified, but the authors stated that statistical analysis (log-rank test) showed a significantly increased risk for these tumour types combined in patients receiving maintenance treatment with hydroxyurea when compared with those under simple surveillance (p = 0.01 or p = 0.03, depending on whether outcomes were analysed according to intention to treat or the main therapy received). The dose of radioactive phosphorus received by the patients who developed leukaemia was moderately higher (0.044 mCi/kg bw per year) than that received by other patients (0.032 mCi/kg bw per year), but the difference was not statistically significant. Seven cases of non-melanoma skin cancer were observed among patients receiving hydroxyurea maintenance and two cases in the surveyed group. [The Working Group noted that the average dose of 0.032 mCi/kg bw per year received by persons without leukaemia does not concord with the average level of 0.021 mCi/kg bw per year for all study subjects followed for more than two years. The Group also noted that the distributions of haematological malignancies by type of treatment were not presented and that the cases of non-Hodgkin lymphoma, chronic lymphocytic leukaemia and multiple myeloma in these elderly people were apparently grouped with the cases of acute myeloid leukaemia and myelodysplastic syndrome before risk analyses were performed, which limits interpretation of the results.]

2.2.2. Essential thrombocythaemia (see also section 2.2.3)

Bellucci et al. (1986) reviewed the medical records of 94 patients (average age, 49.5 years; range, 6–90) in one treatment centre in Paris, France, in whom essential thrombocythaemia had been diagnosed during 1961–82. The patients were followed up for periods ranging from a few months to 19 years, during which time five cases of acute leukaemia were observed. Two of the five cases occurred in the subgroup of 42 patients who had received hydroxyurea as the only chemotherapeutic agent.

In a treatment centre in Limoges, France, Liozon et al. (1997) conducted a retrospective follow-up study of 58 patients (mean age, 66.5 years; range, 18–85) in whom essential thrombocythaemia had been diagnosed during 1981–95. The mean duration of follow-up was approximately five years. Among the 53 patients who had received hydroxyurea as first-line therapy (mean weekly dose, 6 g; range, 2–21 g), one developed acute myeloid leukaemia, one developed chronic myelomonocytic leukaemia and one had myelodysplastic syndrome.

Sterkers et al. (1998) reviewed the medical records of 357 patients (median age, 62 years; range, 30–75) with essential thrombocythaemia diagnosed between 1970 and 1991 and followed-up until 1996 at two haematological centres, in Lille and Lomme, France. Overall, 326 of the 357 patients had been treated with at least one chemotherapeutic agent, and 251 had received hydroxyurea at a starting dose of 1.5 g/day (some had received pipobroman). Within a median duration of follow-up of 8.2 years (range, 1.8–22 years), six patients had developed acute myeloid leukaemia and 11 myelodysplastic syndrome (including chronic myelomonocytic leukaemia). Fourteen of these 17 patients had received hydroxyurea at some time, while seven of the cases were seen in the subgroup of 201 patients who had been treated with hydroxyurea alone.

2.2.3. Chronic myeloproliferative disease

In one treatment centre in Sweden, 81 consecutive patients (age range, 31–82 years) with Philadelphia chromosome-negative chronic myeloproliferative disease, consisting of 35 cases of polycythaemia vera, 32 of essential thrombocythaemia, 12 of myelofibrosis and two of myeloproliferative syndrome [not further specified], were followed prospectively from 1981 to 1989 (Löfvenberg & Wahlin, 1988; Löfvenberg et al., 1990). All had received maintenance treatment with hydroxyurea at a dose of 15–20 mg/kg bw per day. During an average follow-up of 3.9 years (range, one month to 8.7 years), three cases of acute myeloid leukaemia (two cases in patients with essential thrombocythaemia, one in a patient with myelodysplastic syndrome) and one case of myelodysplastic syndrome (in a patient with essential thrombocythaemia) were seen. None of these four cases had been treated with alkylating agents or radioactive phosphorus before treatment with hydroxyurea. No data were available on the number of cancers to be expected among these patients on the basis of incidence rates in the general population.

In another treatment centre in Sweden, Weinfeld et al. (1994) conducted a prospective follow-up of 50 consecutive patients (age range, 33–82 years) with Philadelphia chromosome-negative chronic myeloproliferative disease, consisting of 30 patients with polycythaemia vera, 10 with essential thrombocythaemia and 10 with myelofibrosis, of whom 21, 9 and 7, respectively, had been treated only with hydroxyurea at 60 mg/kg per day for the first week and then 0.5–1.5 g/day. The median observation period was > 10 years, and the minimum was five years, during which time nine cases of acute leukaemia and one case of myelodysplastic syndrome were seen. Seven of the acute leukaemias occurred among 37 patients treated with hydroxyurea only (three in patients with polycythaemia vera, one in a patient with essential thrombocythaemia and three in patients with myelofibrosis), and two of the cases of acute leukaemia and one case of myelodysplastic syndrome occurred among 13 patients treated with alkylating agents prior to entrance into the study, yielding transformation frequencies of [19%] with hydroxyurea and [23%] with previous treatment.

Forty-two patients with polycythaemia vera (16 of whom were treated with hydroxyurea only), 15 with essential thrombocythaemia, six with myelofibrosis with myeloid metaplasia and one with an unclassified myeloproliferative disorder seen at a medical centre in Illinois, USA, during 1993–95 were evaluated for subsequent development of acute leukaemia or myelodysplastic syndrome (Nand et al., 1996). At the date of entry into the study, the patients had survived for a median of 5.2 years (range, four months to 20 years) since diagnosis of their chronic myeloproliferative disease. Five (7.8%) of the 64 cases transformed into acute leukaemia or myelodysplastic syndrome; none of these were in the 11 patients who had received no treatment or aspirin, two (11%) occurred in 18 patients treated with phlebotomy alone, two (8%) in 25 patients who had received hydroxyurea and one (10%) in 10 patients who had received only other immunosuppressive therapy. [The Working Group noted that the study group was composed of prevalent cases of chronic myeloproliferative disease only and that the follow-up period for most patients must have been very short.]

2.2.4. Sickle-cell anaemia

In order to test the efficacy of hydroxyurea in reducing the frequency of painful crises in adults with sickle-cell anaemia, Charache et al. (1995) conducted a randomized, placebo-controlled clinical trial. Of 299 such patients, 152 were assigned to hydroxyurea, while 147 were given placebo. Because of the beneficial effects observed, the trial was stopped after a median of 21 months (range, 14–24 months). At that time, no cases of leukaemia or other neoplastic disorders were seen.

To assess the safety and efficacy of hydroxyurea for the treatment of severe sickle-cell anaemia in children, Scott et al. (1996) conducted a small prospective study of 15 patients in one treatment centre in Illinois, USA. Thirteen patients in whom sickle-cell anaemia had been diagnosed in 1992–95 received hydroxyurea for a median of two years (range, 0.5–3.3 years), during which time no cases of acute leukaemia or other malignancies were seen.

2.2.5. Congenital heart disease

Sixty-four patients ranging in age from 8 to 47 years with inoperable cyanotic congenital heart disease were included in a prospective study in a treatment centre in France (Triadou et al., 1994). The patients received hydroxyurea at an initial dose of 10 mg/kg bw per day, which was adapted according to haematological tolerance and continued over a period ranging from [two to 15 years] (mean, approximately five years). No cases of acute leukaemia or other malignancies were seen.

3. Studies of Cancer in Experimental Animals

The Working Group was aware of early studies in mice (Bhide & Sirsat, 1973) and in rats (Philips & Sternberg, 1975), which were considered inadequate for evaluation.

3.1. Intraperitoneal administration

Groups of 50 mice of each sex of the XVII/G strain were treated intraperitoneally with hydroxyurea [purity not specified] starting at two days of age and then at weekly intervals for one year. The doses per mouse were: 1 mg at two days of age, 3 mg at eight days, 5 mg at 15 days and 10 mg from 30 days to one year of age. One group of 50 mice was kept untreated as controls. The incidences of pulmonary tumours were 30/50 (60%) in control and 16/35 (46%) in treated mice. In a positive control group treated with urethane, 28/30 (93%) of mice had lung tumours (Muranyi-Kovacs & Rudali, 1972).

3.2. Administration with known carcinogens

Groups of 40 female Swiss mice, six to seven weeks of age, received dermal applications of 5 µg of 7,12-dimethylbenz[a]anthracene followed four weeks later by treatment with 1% croton oil for 14 weeks. Hydroxyurea at a dose of 500 mg/kg bw was injected intraperitoneally once at 24 h or twice at 24 and 48 h after the first painting with croton oil. Treatment with two doses of hydroxyurea significantly reduced the incidence of skin papillomas when compared with 7,12-dimethylbenz[a]anthracene and croton oil treatment alone (Chan et al., 1970).

Groups of 16 male and 16 female hairless (hr/hr) Oslo mice were given an intraperitoneal injection of 0 or 5 mg hydroxyurea in 0.5 mL distilled water 30 min before dermal application of 2 mg of N-methyl-N-nitrosourea (MNU). Hydroxyurea enhanced the production of skin tumours by MNU from about 50% to 80%, this effect being attributed to inhibition of DNA synthesis (Iversen, 1982a). No such effect was observed when hydroxyurea was administered simultaneously with or after dermal application of 1 mg of MNU (Iversen, 1982b).

Groups of 36–43 female Wistar rats, weighing about 200 g, were given intraperitoneal injections of hydroxyurea in 23 fractionated consecutive doses of 0.1 mg/kg bw each shortly before and during maximal urothelial cell proliferation (33–55 h after partial cystectomy) produced by MNU administered as a single intravesicular pulse dose of 5 mg/kg bw during the various cell cycle phases. Hydroxyurea inhibited MNU-induced urothelial tumour development, and the degree of this inhibition depended on the cell cycle phase during which MNU was instilled. The numbers of rats with urothelial bladder tumours were: 14/43 in the control group (G0 phase) and 7/37, 4/43 (p < 0.02), 10/46, 10/38, 9/36 and 12/40 in groups receiving MNU during the late G1, early and late S, G2+M, and early and late postmitotic phases, respectively (Kunze et al.,1989).

4. Other Data Relevant to an Evaluation of Carcinogenicity and its Mechanisms

4.1. Absorption, distribution, metabolism and excretion

4.1.1. Humans

Although hydroxyurea has been in clinical use for 30 years, the pharmacokinetics of the compound has been extensively studied only recently. Two useful reviews have been published (Donehower, 1992; Gwilt & Tracewell, 1998), but the best of the limited data available come from the study of Rodriguez et al. (1998). These investigators gave 2 g of hydroxyurea either orally or by intravenous infusion over 30 min in a cross-over design to 29 patients with advanced cancers. They demonstrated clearly that oral and intravenous administration have essentially identical kinetics except for a 19.5% greater maximum plasma concentration (Cmax) after intravenous dosing; the lag time of the peak after oral dosing was 0.22 h. Hydroxyurea is essentially completely absorbed from the human gastrointestinal tract, with a narrow range between subjects. The half-time of hydroxyurea is short, with an initial half-time of 0.63 h after intravenous administration and 1.78 h after oral administration and a terminal half-time of 3.32 h after oral administration and 3.39 h after intravenous administration. The clearance of hydroxyurea given orally or intravenously is identical and rapid, at 76 mL/min per m2, with a mean distribution volume of 19.7 L/m2. In this study, slightly more than one-third of the administered dose was recovered in the urine. The 2-g dose resulted in a mean Cmax of 794 µmol/L after oral administration and 1000 µmol/L after intravenous administration and a mean integrated area under the curve of concentration–time (AUC) of 3600 µmol/L per h after intravenous and 3900 µmol/L per h after oral administration.

Belt et al. (1980) compared oral and intravenous administration of escalating doses of hydroxyurea to patients with advanced malignancies. The maximal tolerated dose was 800 mg/m2 every 4 h by oral administration and 3.0 mg/m2 per min when given intravenously as a continuous 72-h infusion. After oral administration of doses of 500 or 800 mg/m2 every 4 h, the peak concentration in plasma ranged from 5.4 to 24.8 × 10−4 mol/L. The time to attain the peak concentrations was 30–120 min. Two-to threefold variations among patients in the Cmax after oral dosing were found. The Cmax values for continuous intravenous infusion of doses of 2.0–3.5 mg/min per m2 were 5.0–11.5 × 10−4 mol/L. The plasma half-times of hydroxyurea in patients given single oral doses of 400–1200 mg/m2 ranged from 132 to 279 min. In pleural fluid samples obtained from two patients, the concentrations paralleled those found in plasma. The mean half-time after discontinuation of intravenous infusion was about 250 min.

Hydroxyurea enters the cerebrospinal fluid, ascites fluid and serum (Beckloff et al., 1965) and breast milk (Sylvester et al., 1987).

Villani et al. (1996) studied nine HIV-infected patients receiving hydroxyurea at 500 mg twice per day orally with or without zidovudine. The Cmax was 0.135 mmol/L, and the Cmin was 0.0085 mmol/L serum. The rate of clearance was 0.18 L/h per kg bw [12.6 L/h], with a half-time of 2.5 ± 0.5 h. The time to maximum clearance was approximately 0.9 h, and the bioavailability was good.

The maximum tolerated dose in a study of patients with chronic myeloid leukaemia in accelerated phase or blast crisis was 27 g/m2 when given as a 24-h intravenous infusion (Gandhi et al., 1998). Intravenous doses of 8–40 g/m2 resulted in plasma concentrations of 0.9–6.4 mmol/L with a half-time of approximately 3.5 h. A steady state was reached in all patients by 6 h. In this study, the dATP levels in peripheral blast cells decreased by 57%, but DNA synthesis decreased by 80–90%. The concentrations of the other deoxynucleotides were not affected.

About 30–60% of an orally administered dose of hydroxyurea is excreted unchanged by the kidneys (Donehower, 1992), although about 35% is generally excreted (Rodriguez et al., 1998).

Andrae (1984) implicated a cytochrome P450-dependent process in metabolic activation of hydroxyurea which increases its potential for genetic damage. Hydrogen peroxide was reported to be a toxic metabolic product of hydroxyurea (Andrae & Greim, 1979). DeSesso et al. (1994) found that d-mannitol, a scavenger of free radicals, decreases the genotoxic effect of hydroxyurea. Sato et al. (1997) described pathways for the generation of nitric oxide from hydroxyurea via copper-catalysed peroxidation. Hepatic and renal conversion of hydroxyurea by a cytochrome c-dependent pathway to urea may account for 30–50% of administered doses. Urease may degrade hydroxyurea to produce hydroxylamine and ultimately acetohydroxamic acid (Gwilt & Tracewell, 1998).

Both high and low doses show log-linear excretion kinetics, reflecting the predominance of renal mechanisms. The excretion of doses of 10–35 mg/kg bw diverges from linearity, probably because of an increasingly important saturable non-renal metabolic pathway (Villani et al., 1996; Luzzati et al., 1998).

4.1.2. Experimental systems

In contrast to the situation for humans, little information is available on the pharmacokinetics of hydroxyurea in animals, despite its wide use as a model teratogen and to synchronize the cell cycle in cell cultures. The lack of data may be due to the lack of a suitably sensitive assay during the early development of the drug (Donehower, 1992).

Van den Berg et al. (1994) gave nude mice doses of 0–200 mg/kg bw hydroxyurea by intraperitoneal injection and found a plasma concentration of 159 µmol/L within a half-time of only 11 min.

Wilson et al. (1975) found that the half-time of hydroxyurea in rats given 137 mg/kg bw per day intraperitoneally on days 9–12 of gestation was 15 min in the dams and 85 min in the embryos. In rhesus monkeys given 100 mg/kg bw per day intravenously on days 23–32 of gestation, the half-time was 120 min after the last injection in the mothers and 265 min in their fetuses.

4.2. Toxic effects

4.2.1. Humans

The major dose-limiting (and dose-related) toxic effects of hydroxyurea are granulocytopenia, which resolves relatively rapidly after withdrawal of the drug (Belt et al., 1980), and myelosuppression, seen in advanced chronic myeloid leukaemia (Gandhi et al., 1998). Drug-induced dermopathy with characteristics of dermatomyositis have been reported (Richard et al., 1989; Velez et al., 1998) as well as hyperpigmentation of the nails (Gropper et al., 1993; de Montalembert et al., 1997) and leg ulcers (Cox et al., 1997; Weinlich et al., 1998), although one large study of patients with sickle-cell anaemia found an equal rate of leg ulcers in patients given the placebo (Charache et al., 1996). In two- (Charache et al., 1996) and three-year (de Montalembert et al., 1997) follow-up studies of patients with sickle-cell anaemia treated continuously with hydroxyurea, no serious side-effects other than mild neutropenia were observed, and this did not limit treatment.

When very high doses are given intravenously, dose-related mucositis is seen (Gandhi et al., 1998), but neutropenia is usually the treatment-limiting side-effect.

4.2.2. Experimental systems

No formal toxicological studies on hydroxyurea in animals were available to the Working Group.

4.3. Reproductive and prenatal effects

4.3.1. Humans

Information on the use of hydroxyurea in pregnancy is limited to a few case reports. Five case reports involved exposure to hydroxyurea for periods ranging from seven months to four years before pregnancy and throughout gestation at doses of 0.5–3.0 g daily. One woman developed eclampsia at 26 weeks and delivered a stillborn but phenotypically normal infant (Delmer et al., 1992). The other four pregnancies ended in four normal, healthy infants at 36–40 weeks of gestation, with normal blood counts and normal postnatal development up to a maximum of 32 months (Patel et al., 1991; Delmer et al., 1992; Tertian et al., 1992; Jackson et al., 1993). Three other cases have been reported: one woman received a single dose of 8 g of hydroxyurea at about 12 weeks of pregnancy and had an elective termination four weeks later of an apparently normal fetus (Doney et al., 1979). Another woman was treated with an unspecified dose of hydroxyurea for six months before pregnancy and from mid-second trimester to near term, and delivered a healthy infant who developed normally during one year of follow-up (Fitzgerald & McCann, 1993). The third case involved a woman who had been treated with an unspecified dose of hydroxyurea two years before conception. She delivered a normal infant, who had normal physical and mental development at seven years of age (Pajor et al., 1991). [The Working Group noted that the doses of hydroxyurea used clinically are about one-fifth to one-tenth of the teratogenic dose in rodents.]

4.3.2. Experimental systems

Studies on the teratogenicity of hydroxyurea in chicks, mice, rats, rabbits, cats and monkeys have been published since the original reports by Murphy and Chaube (1964) and Chaube and Murphy (1966), who showed that a single intraperitoneal dose of 250 mg/kg bw or more given to Wistar rats on one of days 9–12 of gestation produced a high proportion of fetuses with multiple gross malformations of the central nervous system, palate and skeleton.

Pregnant NMRI mice injected intraperitoneally on day 10 of gestation with 500 mg/kg bw hydroxyurea showed marked necrosis of the neuroepithelium of the spinal cord 4 h after injection. The cytotoxicity could be partially prevented by simultaneous injection of 700 mg/kg bw deoxycytidine monophosphate (Herken, 1984) and completely prevented by simultaneous injection of 1 mg/kg bw colchicine (Herken, 1985). These results suggest that the action of hydroxyurea is dependent both on DNA synthesis and on the cytoskeleton.

Pregnant Wistar-derived rats were dosed intraperitoneally on day 12 of gestation with 250, 500, 750 or 1000 mg/kg bw hydroxyurea, and the fetuses were examined on day 20. A dose-related increase in the frequency of multiple malformations of the viscera and skeleton and reduced fetal weight were observed at doses ≥ 500 mg/kg bw, but embryolethality was seen only at 1000 mg/kg bw. Hydroxyurea was shown to pass into the embryo and to persist there longer than in maternal blood. DNA synthesis, as measured by thymidine incorporation into the embryo, was depressed markedly by doses ≥ 500 mg/kg bw, and marked cytotoxicity was also observed (Scott et al., 1971).

Studies from the same laboratory with the same strain of rat showed that intraperitoneal injection of 375 or 500 mg/kg bw hydroxyurea on day 12 of pregnancy produced microscopic evidence of cytotoxicity in the neural tube, but no malformations were observed when the dams were allowed to deliver their pups at term. Nevertheless, observation of the offspring at 30–50 days of age showed locomotor and behavioural deficits at both doses (Butcher et al., 1973). Further studies from the same laboratory with the same strain of rat showed that teratogenic and embryolethal effects could be induced by a dose as low as 137 mg/kg bw, but not by 100 mg/kg bw, administered intraperitoneally on days 9–12 of gestation (Wilson et al., 1975). Behavioural effects were also observed in the offspring of Sprague-Dawley dams treated with a single intraperitoneal dose of 150 mg/kg bw hydroxyurea on various days of pregnancy (Brunner et al., 1978). The wide range of malformations induced in rats by hydroxyurea has led to its use as a positive control substance in standard testing for both teratogenicity (Aliverti et al., 1980; Price et al., 1985) and developmental toxicity (postnatal behaviour) (Vorhees et al., 1979, 1983). Comparisons of the teratogenic responses in various stocks and strains of rats showed differences in the type of malformation and the time of sensitivity in two stocks of Wistar rats (Barr & Beaudoin, 1981) and in Wistar and Fischer 344 rats (DePass & Weaver, 1982).

A group of 27 pregnant golden hamsters received an intravenous injection of 50 mg/kg bw hydroxyurea on day 8 of pregnancy. The embryos were examined for external malformations only. A high rate of fetal death and malformations, especially of the central nervous system, was observed (Ferm, 1966).

The teratogenicity of hydroxyurea in pregnant New Zealand white rabbits was demonstrated by subcutaneous injection of 750 mg/kg bw once on day 12 of gestation, with embryo and fetal examination 15 min to 32 h later by histology and on day 29 for malformations. Treatment produced marked cytotoxicity and a high percentage of resorptions (61%), reduced fetal weight and malformations in all surviving fetuses affecting most organ systems and the skeleton, as observed in rats (DeSesso & Jordan, 1977; DeSesso, 1981a). The mechanism by which hydroxyurea produces its teratogenic action was investigated in detail by DeSesso and his co-workers, who showed in rabbits that hydroxyurea is not only cytotoxic and inhibits DNA synthesis but also causes a very rapid, marked reduction in uterine–placental blood flow, which may be responsible for some of the teratogenic effects (Millicovsky et al., 1981). In addition, the teratogenic effects can be inhibited by simultaneous administration of the anti-oxidant propyl gallate, which reduces the cytotoxicity (DeSesso, 1981b). This activity occurs within the embryo and is independent of the inhibition of DNA synthesis (DeSesso & Goeringer, 1990). Inhibition of the cytotoxicity and teratogenicity of hydroxyurea by d-mannitol, a potent scavenger of hydroxyl free radicals, suggests that these radicals are the proximate cytotoxins and teratogens (DeSesso et al., 1994).

Groups of 17 mated cats of European and Persian breeds were dosed orally with 50 or 100 mg/kg bw hydroxyurea on days 10–22 of gestation, and the fetuses were examined on day 43. At 50 mg/kg bw, fetal weight and survival were not affected, but a high proportion of the fetuses were malformed, with a wide range of malformations similar to those seen in other species. At 100 mg/kg bw, a large proportion of the cats were not pregnant, but maternal and fetal weights were reduced, the frequency of resorptions increased and one of two live fetuses was malformed (cyclopia) (Khera, 1979).

Of 22 pregnant female rhesus monkeys (Macaca mulatta) dosed intravenously with 50–500 mg/kg bw hydroxyurea for various times between days 18 and 45 of gestation, eight aborted or had intrauterine deaths; 10 had fetuses with multiple malformations mostly of the axial skeleton, but also genitourinary, cardiac, brain, eye and intestinal defects; and the infants of three were growth retarded and one was normal (Theisen et al., 1973; Wilson, 1974; Wilson et al., 1975). [The Working Group noted that little detailed information is given in these reports.]

The teratogenicity of hydroxyurea was compared in mouse embryos in vivo and in vitro, to study the effects of varying the concentration of drug and the duration of exposure. Mated ICR mice were injected intraperitoneally with 300 mg/kg bw hydroxyurea on day 9 of pregnancy (vaginal plug = day 1), and the embryos were removed 48 h later for examination for malformations and for protein content. The embryos of untreated mice were removed on day 9 and cultured in vitro in various concentrations of hydroxyurea for various lengths of time, followed by culture in drug-free medium up to 48 h. In vivo, 45% of the embryos showed malformations, including exencephaly and phocomelia, and the peak plasma concentration of hydroxyurea was 311 ± 22 µg/mL 7 min after injection, with a half-time of 30 min. Culture in vitro with hydroxyurea at 300 µg/mL for 30 min resulted in malformations in 41% of the embryos that were similar to those found in vivo. Culture at a concentration of 500 µg/mL for 30 min or at 250 µg/mL for 1 h resulted in 100% malformed embryos, but culture at 125 µg/mL for 1 h resulted in no malformations (Warner et al., 1983). Culture of 10-day CD rat embryos and eight-day CD-1 mouse embryos with 300 µg/mL hydroxyurea for 1 h followed by 43 h in drug-free medium resulted in impaired development, and the embryos had reduced DNA and protein contents. Addition of various concentrations of dAMP to the culture medium did not inhibit the action of hydroxyurea, and addition of dCMP had minimal inhibitory activity. Hydroxyurea decreased all nucleotide pools, and addition of dAMP increased the pools but not to control levels (Hansen et al., 1995).

Malformations were also produced in chicks injected in ovo on day 4 with 800 µg of hydroxyurea (Iwama et al., 1983).

Seven groups of at least six male C57BL/6J×C3H/HeJ F1 mice were injected when 13–15 weeks of age with 0, 25, 50, 100, 200, 400 or 500 mg/kg bw hydroxyurea intraperitoneally daily for five consecutive days. The epididymides and testes were examined eight and 29 days after the last injection. Body weight was not affected in any of the animals, but the testis weight was reduced in a dose-related manner at all doses except the lowest. A dose-related reduction in DNA synthesis was seen, resulting in depletion of pachytene spermatocytes and a consequent reduction in later cell stages and spermiogenesis. Spermatogonial stem cells were not affected, and showed repopulation of cell stages with normal differentiation kinetics (Evenson & Jost, 1993). Similar results were reported in B6C3/F1/BOM M mice aged six to eight weeks injected intraperitoneally with 200 mg/kg bw hydroxyurea for five days (Wiger et al., 1995).

4.4. Genetic and related effects

4.4.1. Humans

In studies of genetic alterations in leukaemic cells of patients treated with hydroxyurea, a statistically non-significant association was seen between treatment with hydroxyurea alone or in combination and the occurrence of leukaemia and myelodysplastic syndrome characterized by abnormalities of chromosome 17 in patients with essential thrombocythaemia [p = 0.11, Fisher’s exact test]. As discussed in section 2, Sterkers et al. (1998) monitored the occurrence of acute leukaemia and myelodysplastic syndrome in 251 patients with essential thrombocythemia who were treated with hydroxyurea. The findings in the leukaemic cells are summarized in Table 2. In seven cases of leukaemia treated with hydroxyurea, including three given the drug alone, there were rearrangements of chromosome 17, including unbalanced translocations, partial or complete deletions and isochromosome 17q, which resulted in 17p deletion in the leukaemic cells. P53 mutation was observed in six cases, including two treated with hydroxyurea alone. The authors suggested that the molecular characteristics of these leukaemias are consistent with 17p syndrome and that prolonged use of hydroxyurea in patients with essential thrombocythaemia may lead to acute myeloid leukaemia and myelodysplastic syndrome with loss of chromosome 17p material and P53 mutation. A review of the literature by these authors revealed similar 17p deletions in four of 11 patients treated for essential thrombocythaemia with hydroxyurea alone but in only one of 24 patients who did not receive this treatment. Tefferi (1998) cautioned, however, that the results of bone-marrow and cytogenetic investigations before treatment were not available for some of the patients. Monosomy 17 was also observed in complex karyotypes in two of three cases of leukaemia reported by Liozon et al. (1997) among 58 patients with essential thrombocythaemia treated with hydroxyurea; in the third case, which was chronic myelomonocytic leukaemia, the karyotype was normal.

Table 2. Karyotypic findings in the bone marrow of patients with essential thrombocythaemia treated with hydroxyurea.

Table 2

Karyotypic findings in the bone marrow of patients with essential thrombocythaemia treated with hydroxyurea.

Quesnel et al. (1993) identified the t(8;21) translocation in a case of leukaemia in which essential thrombocythaemia had been treated with hydroxyurea alone. The t(8;21) is associated with the French–American–British M2 (acute myeloblastic) subtype of denovo and treatment-related acute myeloid leukaemia. The complex karyotype, which also contained monosomy 17, was 46, XX[3]/47, XX, +8 [2]/43–44,XX,der(7)t(7;dup5), t(8;21)(q22;q22), −16,−17,t(18;?)(q;?)[10].

Ören et al. (1999) described a case of acute promyelocytic leukaemia with i(17q) after treatment of Philadelphia chromosome-positive chronic myeloid leukaemia with combination therapy including hydroxyurea, but i(17q) may occur in chronic myeloid leukaemia in blast crisis.

Diverse chromosomal aberrations have been seen in human bone-marrow cells after hydroxyurea treatment. Diez-Martin et al. (1991) reviewed studies of the chromosomes of 104 patients at various stages of polycythaemia vera. The bone-marrow cells of five of six patients treated with hydroxyurea alone had abnormalities, including an unbalanced t(1;7)(p11;p11), which can be associated with treatment-related myelodysplastic syndrome, but this abnormality may occur without prior treatment. Cytogenetic analyses in these five patients were performed only on bone-marrow samples obtained after treatment. One each of the other four abnormal marrows had t(8;13)(p21;q12), +9, del(6)(q13q21) and t(1;?)(q12;?). Furthermore, the authors observed several de-novo abnormalities in untreated patients which they related to the disease itself rather than to the therapy, including +9, +8 and 20q–, and suggested that the 13q– abnormality is related to disease progression.

Löfvenberg et al. (1990) examined 81 hydroxyurea-treated patients with Philadelphia chromosome-negative chronic myeloproliferative disorders, comprising 35 with polycythaemia vera, 32 with essential thrombocythaemia, 12 with myelofibrosis and two with myeloproliferative syndromes. Only three had received prior therapy with alkylating agents or radioactive phosphorus. Four of the 81 developed acute myeloid leukaemia or myelodysplastic syndrome. Five of 53 evaluable patients (9%) had clonal cytogenetic abnormalities involving chromosomes 1, 9, 20 and 21 before treatment, and 15% had these abnormalities at follow-up, during or after hydroxyurea treatment. Treatment was thus associated with a low frequency of cytogenetic abnormalities in a heterogeneous population, and the abnormalities observed before and after treatment were similar.

The series later reported on by Weinfeld et al. (1994) included 30 patients with polycythaemia vera, 10 with essential thrombocythaemia and 10 with myelofibrosis who were treated with hydroxyurea. Acute leukaemia developed in nine patients and myelodysplastic syndrome in one; seven of the leukaemia patients had been treated with hydroxyurea alone. The duration of therapy for patients who developed leukaemia or myelodysplastic syndrome was 5–111 months. Seven of 19 previously untreated patients with initially normal karyotypes treated with hydroxyurea alone developed clonal chromosomal abnormalities during therapy (37%).

Davidovitz et al. (1998) observed evolution of polycythaemia vera to myelofibrosis with a t(1;20)(q32;q13.3) in a patient who received chronic low-dose hydroxyurea. The t(1;20) affected the same region of chromosome 20 as the 20q– abnormality; it could not be determined whether the translocation was related to the treatment. Furgerson et al. (1996) described a patient in whom essential thrombocythaemia evolved to acute myeloid leukaemia after hydroxyurea treatment. The karyotype was normal at the time of diagnosis of essential thrombocythaemia but revealed del(5)(q23), del(7)(q31), inv(16)(p13;q22),+8 when acute myeloid leukaemia emerged.

4.4.2. Experimental systems

Early studies on the mutagenicity of hydroxyurea were summarized by Timson (1975). Reviews on the mutagenicity of anticancer drugs in general, including hydroxyurea, were provided by Ferguson (1995) and Jackson et al. (1996). Ferguson and Denny (1995) commented on some practical issues in testing antimetabolites, which may limit the usefulness (and meaning) of some types of in-vitro assays.

The results of tests for genotoxicity with hydroxyurea are summarized in Table 3.

Table 3. Genetic and related effects of hydroxyurea.

Table 3

Genetic and related effects of hydroxyurea.

Hydroxyurea was inactive as either a frameshift or base-pair substitution mutagen in Salmonella typhimurium strains TA1537, TA1535, TA98 and TA100, and addition of an exogenous metabolic activation system did not affect these results. Hydroxyurea induced SOS repair in Escherichia coli K12 cells. In various Saccharomyces cerevisiae strains, hydroxyurea induced mitotic crossing over, mitotic gene conversion, intrachromosomal recombination and aneuploidy, but not ‘petite’ mutations. It also increased the frequency of ultraviolet-induced mitotic gene conversion and induced recombination in dividing but not G1 or G2 arrested cells of the RS112 strain of yeast. In meiotic yeast cells, hydroxyurea increased the frequency of meiotic recombination.

Hydroxyurea is a clastogen in mammalian cells in vitro and in vivo.

Hydroxyurea caused chromosomal aberrations in cultured Chinese hamster cells, in mouse cells and in various human cell lines. Karon and Benedict (1972) found that hydroxyurea induced chromosomal aberrations when given during S phase but not when given during G2 phase. It did not induce micronuclei in human peripheral blood lymphocytes but increased the frequency of sister chromatid exchange and of gene amplification in L5178Y mouse lymphoma cells. Hydroxyurea induced sister chromatid exchange in various Chinese hamster cell lines in vitro. It caused DNA strand breaks in Ehrlich ascites tumour cells and in human T lymphoma cells in vitro.

It did not induce mutations at the HPRT locus in a cultured human T-lymphocyte cell line at doses of 50–250 µmol/L, which had substantial effects on DNA synthesis, but induced mutants at the Tk locus in L5178Y cells.

Hydroxyurea caused cell transformation in mass cultures of embryonic cells from BN/a, mice, but not in cultures derived from two other strains of mice—DBA/2 and Swiss, nor in BALB/c 3T3 cells. Although hydroxyurea alone did not induce morphological transformation in Syrian hamster embryo cells, the cell cycle arrest caused by the drug led to enhancement of cell transformation by bromodeoxyuridine (Tsutsui et al., 1979). Hydroxyurea did not enhance metabolic cooperation between V79 cells (Toraason et al., 1992).

Hydroxyurea treatment led to hypermethylation of DNA in hamster fibrosarcoma cells (Nyce et al., 1986). In rats, this resulted in nitric oxide production (Jiang et al., 1997) and induced a cytokine response (Navarra et al., 1995, 1997). These effects may indicate an enhanced effect on chromosomal damage in certain situations in vivo. Hydroxyurea also induced DNA hypermethylation in normal human embryonic lung fibroblasts (WI-38) and their simian virus 40-transformed counterparts (SVWI-38) (De Haan & Parker, 1988).

Hydroxyurea induced micronuclei in the bone marrow of non-tumour-bearing male NMRI mice but did not induce micronucleated cells in female C57BL/6 × C3H/He hybrid mice, although it produced sperm abnormalities in male mice of this strain.

Although hydroxyurea is a mutagen in somatic cells, there is no evidence that it mutates germ cells. It did not cause dominant lethal mutation or specific locus mutation in mice. It did not induce chromosomal damage in spermatogonial cells of male Swiss mice, although it enhanced damage induced by X-rays.

Minford et al. (1984) showed that hydroxyurea at 0.1 mmol/L enhanced both DNA breakage and cytotoxicity caused by the intercalating DNA topoisomerase II inhibitor, amsacrine. Lambert et al. (1983) found similar results in relation to adriamycin. Palitti et al. (1984a) showed that treatment with hydroxyurea after mitomycin C enhanced the frequencies of chromosomal aberrations and sister chromatid exchange induced by mitomycin C alone in both Chinese hamster cells and human lymphocytes. Hydroxyurea had a synergistic effect on ultraviolet-induced sister chromatid exchange (Ishii & Bender, 1980) and enhanced X-radiation-induced damage in spermatogonial cells of Swiss mice (van Buul & Bootsma, 1994).

4.5. Mechanistic considerations

Hydroxyurea does not bind or bond to DNA but acts by inhibiting ribonucleotide reductase, which converts ribonucleoside diphosphates to deoxyribonucleotide diphosphates, the precursers for de-novo DNA synthesis. Hydroxyurea depletes intracellular deoxyribonucleotide pools and is known and used as an inhibitor of DNA synthesis (Timson, 1975).

The differences in the results of various studies may depend on the exact cell culture conditions, especially in regard to the amounts of deoxyribonucleotides available. Hansen et al. (1995) found that they could partially attenuate the embryotoxic effects of hydroxyurea by providing additional deoxyribonucleotides.

In a number of experiments, hydroxyurea appeared to enhance the susceptibility of cells to mutagenesis by other agents (e.g. Palitti et al., 1983, 1984a,b; Ferguson, 1990; Jelmert et al., 1992). There are three possible reasons for this:

  1. It halts the progression of cells in the late G1 phase of the cycle, allowing synchronization of the culture (e.g. Tsutsui et al., 1979). Thus, if cells are sensitive to a certain agent in a particular phase of the cell cycle, hydroxyurea may reveal this effect.
  2. Although hydroxyurea inhibits normal DNA synthesis, it does not appear to inhibit unscheduled DNA synthesis at the same doses after treatment with various genotoxic agents, including X-rays (e.g. Painter & Cleaver, 1967). This justifies its inclusion in protocols of unscheduled DNA synthesis.
  3. Prempree and Merz (1969) suggested that hydroxyurea could inhibit the repair of chromosomal breaks without itself inducing breaks.

5. Summary of Data Reported and Evaluation

5.1. Exposure data

Hydroxyurea is a chemically simple antimetabolite that inhibits the enzyme ribonucleotide reductase. It has been in clinical use since the 1960s and is widely used for the treatment of severe sickle-cell disease, chronic myeloid leukaemia, myeloproliferative disorders such as polycythaemia vera and essential thrombocythaemia and, increasingly, in combination with didanosine in HIV infection. Hydroxyurea is sometimes used for the treatment of psoriasis and various solid tumours.

5.2. Human carcinogenicity data

The risk for leukaemia associated with administration of hydroxyurea in the treatment of chronic myeloproliferative disorders has been evaluated in a number of small cohort studies. Overall, 5–6% of patients developed either acute leukaemia or myelodysplastic syndrome subsequent to the start of hydroxyurea treatment. Large variation in the length of active follow-up was not taken into account in the analyses. The risk for leukaemia in patients with chronic myeloproliferative disorders who were not treated with hydroxyurea or other agents (e.g. polycythaemia vera patients treated with phlebotomy alone) was also increased in comparison with that of the general population. The available data do not allow a conclusion about whether the occurrence of acute leukaemia and myelodysplastic syndrome in the hydroxyurea-treated patients represents progression of the myeloproliferative process or an effect of the treatment.

5.3. Animal carcinogenicity data

Hydroxyurea was tested in one experiment in mice by intraperitoneal administration beginning at two days of age. No increase in the incidence of tumours was reported. Hydroxyurea has also been tested in combination with other chemical carcinogens to assess the effect of inhibition of DNA synthesis on carcinogenesis. The experiments are inadequate to assess the carcinogenicity of hydroxyurea.

5.4. Other relevant data

Hydroxyurea is readily absorbed after oral administration. In one study, 35% of an administered dose was excreted unchanged in the urine of humans. Hydroxyurea is widely distributed in tissues. Its main toxic effect is neutropenia.

Hydroxyurea is teratogenic and causes postnatal behavioural deficits after prenatal exposure in all species of animals in which it has been tested. It has commonly been used as positive control substance in studies of developmental toxicity.

In one study of patients treated with hydroxyurea for essential thrombocythaemia who developed leukaemia, a statistically non-significant association was found with a 17p chromosomal deletion in leukaemic cells.

Hydroxyurea neither bonds chemically nor otherwise binds to DNA. Instead, it inhibits ribonucleotide reductase, which converts ribonucleoside diphosphates to deoxyribonucleotide diphosphate precursers for de-novo DNA synthesis. Hydroxyurea does not induce gene mutation in bacteria and does not cause mutation at the Hprt locus in mammalian cells. It causes chromosomal mutations and mutagenic effects at the Tk locus in mouse lymphoma cells. It is an effective recombinogen in yeast and induces sister chromatid exchange in mammalian cells. It also causes gene amplification in mammalian cells and may lead to transformation of some but not all cell lines. Although it has been reported to be ineffective in causing germ-cell mutation, it has not been extensively tested for that end-point.

5.5. Evaluation

There is inadequate evidence in humans for the carcinogenicity of hydroxyurea.

There is inadequate evidence in experimental animals for the carcinogenicity of hydroxyurea.

Overall evaluation

Hydroxyurea is not classifiable as to its carcinogenicity to humans (Group 3).

6. References

  • Aliverti V., Bonanomi L., Giavini E. Hydroxyurea as a reference standard in teratological screening. Comparison of the embryotoxic and teratogenic effects following single intraperitoneal or repeated oral administrations to pregnant rats. Arch. Toxicol. 1980 Suppl. 4:239–247. [PubMed: 6933910]
  • Amacher D.E., Turner G.N. The mutagenicity of 5-azacytidine and other inhibitors of replicative DNA synthesis in the L5178Y mouse lymphoma cell. Mutat. Res. 1987;176:123–131. [PubMed: 2432424]
  • American Hospital Formulary Service (1997) AHFS Drug Information® 97, Bethesda, MD, American Society of Health-System Pharmacists, pp. 766–770.
  • Andrae U. Evidence for the involvement of cytochrome P-450-dependent monooxygenase(s) in the formation of genotoxic metabolites from N-hydroxyurea. Biochem. biophys. Res. Commun. 1984;118:409–415. [PubMed: 6704084]
  • Andrae U., Greim H. Induction of DNA repair replication by hydroxyurea in human lymphoblastoid cells mediated by liver microsomes and NADPH. Biochem. biophys. Res. Commun. 1979;87:50–58. [PubMed: 36891]
  • van den Anker-Lugtenburg P.J., Sizoo W. Myelodysplastic syndrome and secondary acute leukemia after treatment of essential thrombocythemia with hydroxyurea. Am. J. Hematol. 1990;33:152. [PubMed: 2301374]
  • Banga S.S., Shenkar R., Boyd J.B. Hypersensitivity of Drosophila mei-41 mutants to hydroxyurea is associated with reduced mitotic chromosome stability. Mutat. Res. 1986;163:157–165. [PubMed: 3093854]
  • Barbé J., Villaverde A., Guerrero R. Induction of the SOS response by hydroxyurea in Escherichia coli K12. Mutat. Res. 1987;192:105–108. [PubMed: 3309645]
  • Barr M. Jr, Beaudoin A.B. An exploration of the role of hydroxyurea injection time in fetal growth and terartogenesis in rats. Teratology. 1981;24:163–167. [PubMed: 7336359]
  • Beckloff G.L., Lerner H.J., Frost D., Russo-Alesi F.M., Gitomer S. Hydroxyurea in biologic fluids: Dose–concentration relationship. Cancer Chemother. Rep. 1965;48:57–58. [PubMed: 5834739]
  • Bellucci S., Janvier M., Tobelem G., Flandrin G., Charpak Y., Berger R., Boiron M. Essential thrombocythemias. Clinical evolutionary and biological data. Cancer. 1986;58:2440–2447. [PubMed: 3768838]
  • Belt R.J., Haas C.D., Kennedy J., Taylor S. Studies of hydroxyurea administered by continuous infusion: Toxicity, pharmacokinetics, and cell synchronization. Cancer. 1980;46:455–462. [PubMed: 7397621]
  • Best P.J.M., Petitt R.M. Multiple skin cancers associated with hydroxyurea therapy. Mayo Clin. Proc. 1998;73:961–963. [PubMed: 9787746]
  • Bhide S.V., Sirsat M.V. Delayed effects of a single treatment of hydroxyurea to newborn mice. Indian J. Cancer. 1973;10:26–30. [PubMed: 4351637]
  • Biron F., Lucht F., Peyramond D., Fresard A., Vallet T., Nugier F., Grange J., Malley S., Hamedi-Sangsari F., Vila J. Pilot clinical trial of the combination of hydroxyurea and didanosine in HIV-1 infected individuals. Antiviral Res. 1996;29:111–113. [PubMed: 8721560]
  • Boyd A.S., Neldner K.H. Hydroxyurea therapy. J. am. Acad. Dermatol. 1991;25:518–524. [PubMed: 1918491]
  • British Medical Association/Royal Pharmaceutical Society of Great Britain (1998) British National Formulary, No. 36, London, p. 377.
  • British Pharmacopoeial Commission (1993) British Pharmacopoeia 1993, London, Her Majesty’s Stationery Office, Vols I & II, pp. 343, 954, S68.
  • Bruce W.R., Heddle J.A. The mutagenic activity of 61 agents as determined by the micronucleus, Salmonella, and sperm abnormality assays. Can. J. Genet. Cytol. 1979;21:319–334. [PubMed: 393369]
  • Brunner R.L., McLean M., Vorhees C.V., Butcher R.E. A comparison of behavioral and anatomical measures of hydroxyurea induced abnormalities. Teratology. 1978;18:379–384. [PubMed: 741390]
  • Budavari, S., ed. (1996) The Merck Index, 12th Ed., Whitehouse Station, NJ, Merck & Co., p. 833.
  • Butcher R.E., Scott W.J., Kazmaier K., Ritter E.J. Postnatal effects in rats of prenatal treatment with hydroxyurea. Teratology. 1973;7:161–165. [PubMed: 4725545]
  • van Buul P.P., Bootsma A.L. The induction of chromosomal damage and cell killing in mouse spermatogonial stem cells following combined treatments with hydroxyurea, 3-aminobenzamide and X-rays. Mutat. Res. 1994;311:217–224. [PubMed: 7526186]
  • Callot-Mellot C., Bodemer C., Chosidow O., Frances C., Azgui Z., Varet B., de Prost Y. Cutaneous carcinoma during long-term hydroxyurea therapy: A report of 5 cases. Arch. Dermatol. 1996;132:1395–1397. [PubMed: 8915331]
  • Cattanach B.M., Peters J., Rasberry C. Induction of specific locus mutations in mouse spermatogonial stem cells by combined chemical X-ray treatments. Mutat. Res. 1989;212:91–101. [PubMed: 2725545]
  • Chan P.C., Goldman A., Wynder E.L. Hydroxyurea: Suppression of two-stage carcinogenesis in mouse skin. Science. 1970;168:130–132. [PubMed: 5417054]
  • Charache S., Terrin M.L., Moore R.D., Dover G.J., Barton F.B., Eckert S.V., McMahon R.P., Bonds D.R. the Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia, author. Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. New Engl. J. Med. 1995;332:1317–1322. [PubMed: 7715639]
  • Charache S., Barton F.B., Moore R.D., Terrin M.L., Steinberg M.H., Dover G.J., Ballas S.K., McMahon R.P., Castro O., Orringer E.P. the Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia, author. Hydroxyurea and sickle cell anemia. Clinical utility of a myelosuppressive ‘switching’ agent. Medicine. 1996;75:300–326. [PubMed: 8982148]
  • Chaube S., Murphy M.L. The effects of hydroxyurea and related compounds on the rat fetus. Cancer Res. 1966;26:1448–1457. [PubMed: 5911587]
  • Chlopkiewicz B., Koziorowska J.H. Transforming activities of methotrexate, hydroxyurea and 5-fluorouracil in different cell systems. Neoplasma. 1983;30:295–302. [PubMed: 6866161]
  • CIS Information Services (1998) Worldwide Bulk Drug Users Directory 1997/98 Edition, Dallas, TX [CD-ROM]
  • Cox C., Nowicky D., Young R. Hydroxyurea-related ankle ulcers in patients with myeloproliferative disorders: A case report and review of the literature. Ann. plast. Surg. 1997;39:546–549. [PubMed: 9374154]
  • Currie J.L., Blessing J.A., Muss H.B., Fowler J., Berman M., Burke T.W. Combination chemotherapy with hydroxyurea, dacarbazine (DTIC), and etoposide in the treatment of uterine leiomyosarcoma: A Gynecologic Oncology Group study. Gynecol. Oncol. 1996a;61:27–30. [PubMed: 8626112]
  • Currie J.L., Blessing J.A., McGehee R., Soper J.T., Berman M. Phase II trial of hydroxyurea, dacarbazine (DTIC), and etoposide (VP-16) in mixed mesodermal tumors of the uterus: A Gynecologic Oncology Group study. Gynecol. Oncol. 1996b;61:94–96. [PubMed: 8626125]
  • Davidovitz Y., Lev D., Ballin A., Tsudik A., Meytes D. Short communication. Translocation (1;20)(q32;q13.3) in myelofibrosis following polycythemia vera. Cancer Genet. Cytogenet. 1998;101:156–158. [PubMed: 9494620]
  • De Boer R.J., Boucher C.A., Perelson A.S. Target cell availability and the successful suppression of HIV by hydroxyurea and didanosine. AIDS. 1998;12:1567–1570. [PubMed: 9764774]
  • De Haan J.B., Parker M.I. Differential effects of DNA synthesis inhibitors on DNA methylation in normal and transformed cells. Anticancer Res. 1988;8:617–620. [PubMed: 2845852]
  • Delmer A., Rio B., Bauduer F., Ajchenbaum F., Marie J.-P., Zittoun R. Pregnancy during myelosuppressive treatment for chronic myelogenous leukaemia (Letter to the Editor). Br. J. Haematol. 1992;82:783–784. [PubMed: 1482675]
  • DePass L.R., Weaver E.V. Comparison of teratogenic effects of aspirin and hydroxyurea in the Fischer 344 and Wistar strains. J. Toxicol. environ. Health. 1982;10:297–305. [PubMed: 7143483]
  • DeSesso J.M. Comparative ultrastructural alterations in rabbit limb-buds after a teratogenic dose of either hydroxyurea or methotrexate. Teratology. 1981a;23:197–215. [PubMed: 7268637]
  • DeSesso J.M. Amelioration of teratogenesis. I. Modification of hydroxyurea-induced teratogenesis by the antioxidant propyl gallate. Teratology. 1981b;24:19–35. [PubMed: 7302871]
  • DeSesso J.M., Goeringer G.C. The nature of the embryo-protective interaction of propyl gallate with hydroxyurea. Reprod. Toxicol. 1990;4:145–152. [PubMed: 2136029]
  • DeSesso J.M., Jordan R.L. Drug-induced limb dysplasias in fetal rabbits. Teratology. 1977;15:199–211. [PubMed: 867277]
  • DeSesso J.M., Scialli A.R., Goeringer G.C. d-Mannitol, a specific hydroxyl free radical scavenger, reduces the developmental toxicity of hydroxyurea in rabbits. Teratology. 1994;49:248–259. [PubMed: 8073363]
  • De Simone C., Guerriero C., Guidi B., Rotoli M., Venier A., Tartaglione R. Multiple squamous cell carcinomas of the skin during long-term treatment with hydroxyurea. Eur. J. Dermatol. 1998;8:114–115. [PubMed: 9649662]
  • Diez-Martin J.L., Graham D.L., Petitt R.M., Dewald G.W. Chromosome studies in 104 patients with polycythemia vera. Mayo Clin. Proc. 1991;66:287–299. [PubMed: 2002687]
  • Disdier P., Harle J.R., Grob J.J., Weiller-Merli C., Magalon G., Weiller P.J. Rapid development of multiple squamous-cell carcinomas during chronic granulocytic leukemia. Dermatologica. 1991;183:47–48. [PubMed: 1769415]
  • Donehower R.C. An overview of the clinical experience with hydroxyurea. Semin. Oncol. 1992;19 Suppl. 9:11–19. [PubMed: 1641651]
  • Doney K.C., Kraemer K.G., Shepard T.H. Combination chemotherapy for acute myelocytic leukemia during pregnancy: Three case reports. Cancer Treat. Rep. 1979;63:369–371. [PubMed: 284844]
  • Donovan P.B., Kaplan M.E., Goldberg J.D., Tatarsky I., Najean Y., Silberstein E.B., Knospe W.H., Laszlo J., Mack K., Berk P.D., Wasserman L.R. Treatment of polycythemia vera with hydroxyurea. Am. J. Hematol. 1984;17:329–334. [PubMed: 6496458]
  • Editions du Vidal (1998) Dictionnaire Vidal 1998, 74th Ed., Paris, OVP, pp. 889–890.
  • Epstein S.S., Arnold E., Andrea J., Bass W., Bishop Y. Detection of chemical mutagens by the dominant lethal assay in the mouse. Toxicol. appl. Pharmacol. 1972;23:288–325. [PubMed: 5074577]
  • Evenson D.P., Jost L.K. Hydroxyurea exposure alters mouse testicular kinetics and sperm chromatin structure. Cell Prolif. 1993;26:147–159. [PubMed: 8471672]
  • Fenech M., Rinaldi J., Surralles J. The origin of micronuclei induced by cytosine arabinoside and its synergistic interaction with hydroxyurea in human lymphocytes. Mutagenesis. 1994;9:273–277. [PubMed: 7934968]
  • Ferguson L.R. Mutagenic and recombinogenic consequences of DNA-repair inhibition during treatment with 1,3-bis(2-chloroethyl)-1-nitrosourea in Saccharomyces cerevisiae. Mutat. Res. 1990;241:369–377. [PubMed: 2198466]
  • Ferguson, L.R. (1995) Mutagenic properties of anticancer drugs. In: Ponder, B.A.J. & Waring, M.J., eds, The Genetics of Cancer, Lancaster, Kluwer Academic Publisher, pp. 177–216.
  • Ferguson L.R., Denny W.A. Anticancer drugs: An underestimated risk or an under-utilised resource in mutagenesis. Mutat. Res. 1995;331:1–26. [PubMed: 7666858]
  • Ferguson L.R., Turner P.M. ‘Petite’ mutagenesis by anticancer drugs in Saccharomyces cerevisiae. Eur. J. Cancer clin. Oncol. 1988a;24:591–596. [PubMed: 3289944]
  • Ferguson L.R., Turner P.M. Mitotic crossing-over by anticancer drugs in Saccharomyces cerevisiae strain D5. Mutat. Res. 1988b;204:239–249. [PubMed: 3278216]
  • Ferm V.H. Severe developmental malformations. Arch. Pathol. 1966;81:174–177.
  • Ferster A., Vermylen C., Cornu G., Buyse M., Corazza F., Devalck C., Fondu P., Toppet M., Sariban E. Hydroxyurea for treatment of severe sickle cell anemia: A pediatric clinical trial. Blood. 1996;88:1960–1964. [PubMed: 8822914]
  • Fibach E., Burke K.P., Schechter A.N., Noguchi C.T., Rodgers G.P. Hydroxyurea increases fetal hemoglobin in cultured erythroid cells derived from normal individuals and patients with sickle cell anemia or beta-thalassemia. Blood. 1993;81:1630–1635. [PubMed: 7680923]
  • Fitzgerald J.M., McCann S.R. The combination of hydroxyurea and leucapheresis in the treatment of chronic myeloid leukaemia in pregnancy. Clin. Lab. Haematol. 1993;15:63–65. [PubMed: 8472499]
  • Foli A., Maserati R., Minoli L., Wainberg M.A., Gallo R.C., Lisziewicz J., Lori F. Therapeutic advantage of hydroxyurea and didanosine combination therapy in patients previously treated with zidovudine. AIDS. 1998;12:1113–1114. [PubMed: 9662215]
  • Furgerson J.L., Vukelja S.J., Baker W.J., O’Rourke T.J. Acute myeloid leukemia evolving from essential thrombocythemia in two patients treated with hydroxyurea. Am. J. Hematol. 1996;51:137–140. [PubMed: 8579054]
  • Galli A., Schiestl R.H. Hydroxyurea induces recombination in dividing but not in G1 or G2 cell cycle arrested yeast cells. Mutat. Res. 1996;354:69–75. [PubMed: 8692208]
  • Gandhi V., Plunkett W., Kantarjian H., Talpaz M., Robertson L.E., O’Brien S. Cellular pharmacodynamics and plasma pharmacokinetics of parenterally infused hydroxyurea during a phase I clinical trial in chronic myelogenous leukemia. J. clin. Oncol. 1998;16:2321–2331. [PubMed: 9667246]
  • Gao W.Y., Zhou B.S., Johns D.G., Mitsuya H., Yen Y. Role of the M2 subunit of ribonucleotide reductase in regulation by hydroxyurea of the activity of the anti-HIV-1 agent 2′,3′-dideoxyinosine. Biochem. Pharmacol. 1998;56:105–112. [PubMed: 9698094]
  • Gennaro, A.R. (1995) Remington: The Science and Practice of Pharmacy, 19th Ed., Easton, PA, Mack Publishing Co., Vol. II, p. 1252.
  • Graham, P.J. (1955) Synthesis of Ureas. US Patent No. 2,705,727. Assigned to E.I. du Pont de Nemours & Co., Wilmington, DE.
  • Greenwood S.K., Armstrong M.J., Hill R.B., Bradt C.I., Johnson T.E., Hilliard C.A., Galloway S.M. Fewer chromosome aberrations and earlier apoptosis induced by DNA synthesis inhibitors, a topoisomerase II inhibitor or alkylating agents in human cells with normal compared with mutant p53. Mutat. Res. 1998;401:39–53. [PubMed: 9639670]
  • Gropper C.A., Don P.C., Sadjadi M.M. Nail and skin hyperpigmentation associated with hydroxyurea therapy for polycythemia vera. Int. J. Dermatol. 1993;32:731–733. [PubMed: 8225715]
  • Guilhot F., Abgrall J.-F., Harousseau J.-L., Bauters F., Brice P., Dine G., Tilly H., Ifrah N., Cassasus P., Rochant H., Christian B., Guerci A., Lamagnere J.-P., Le Prise P.-Y., Duclos B., Tanzer J. A multicentric randomised study of alpha 2b interferon (IFN) and hydroxyurea (HU) with or without cytosine-arabinoside (Ara-c) in previously untreated patients with Ph+ chronic myelocytic leukemia (CML): Preliminary cytogenetic results. Leuk. Lymphoma. 1993;11 Suppl. 1:181–183. [PubMed: 8251893]
  • Gwilt P.R., Tracewell W.G. Pharmacokinetics and pharmacodynamics of hydroxyurea. Clin. Pharmacokinet. 1998;34:347–358. [PubMed: 9592619]
  • Hahn P., Kapp L.N., Morgan W.F., Painter R.B. Chromosomal changes without DNA overproduction in hydroxyurea-treated mammalian cells: Implications for gene amplification. Cancer Res. 1986;46:4607–4612. [PubMed: 3731112]
  • Hansen D.K., Grafton T.F., Cross D.R., James S.J. Partial attenuation of hydroxyurea-induced embryotoxicity by deoxyribonucleotides in mouse and rat embryos treated in vitro. Toxicol. in Vitro. 1995;9:11–19. [PubMed: 20650058]
  • Hart J.W., Hartley-Asp B. Induction of micronuclei in the mouse. Revised timing of the final stage of erythropoiesis. Mutat. Res. 1983;120:127–132. [PubMed: 6843575]
  • Haworth S., Lawlor T., Mortelmans K., Speck W., Zeiger E. Salmonella mutagenicity test results for 250 chemicals. Environ. Mutag. 1983;5 Suppl. 1:3–142. [PubMed: 6365529]
  • Herken R. The influence of deoxycytidine monophosphate (dCMP) on the cytotoxicity of hydroxyurea in the embryonic spinal cord of the mouse. Teratology. 1984;30:83–90. [PubMed: 6484855]
  • Herken R. Ultrastructural changes in the neural tube of 10-day-old mouse embryos exposed to colchicine and hydroxyurea. Teratology. 1985;31:345–352. [PubMed: 4012644]
  • Hill A.B., Schimke R.T. Increased gene amplification in L5178Y mouse lymphoma cells with hydroxyurea-induced chromosomal aberrations. Cancer Res. 1985;45:5050–5057. [PubMed: 4027984]
  • Holcombe R.F., Treseler P.A., Rosenthal D.S. Chronic myelomonocytic leukemia transformation in polycythemia vera. Leukemia. 1991;5:606–610. [PubMed: 2072746]
  • Ishii Y., Bender M.A. Effects of inhibitors of DNA synthesis on spontaneous and ultraviolet light-induced sister-chromatid exchanges in Chinese hamster cells. Mutat. Res. 1980;79:19–32. [PubMed: 6448958]
  • Iversen O.H. Enhancement of methylnitrosourea skin carcinogenesis by inhibiting cell proliferation with hydroxyurea or skin extracts. Carcinogenesis. 1982a;3:881–889. [PubMed: 7127669]
  • Iversen O.H. Hydroxyurea enhances methylnitrosourea skin carcinogenesis when given shortly before, but not after, the carcinogen. Carcinogenesis. 1982b;3:891–894. [PubMed: 7127670]
  • Iwama M., Sakamoto Y., Honda A., Mori Y. Limb deformity induced in chick embryo by hydroxyurea. J. Pharmacobiodyn. 1983;6:836–843. [PubMed: 6668544]
  • Jackson H., Shukri A., Ali K. Hydroxyurea treatment for chronic myeloid leukaemia during pregnancy. Br. J. Haematol. 1993;85:203–204. [PubMed: 8251394]
  • Jackson M.A., Stack H.F., Waters M.D. Genetic activity profiles of anticancer drugs. Mutat. Res. 1996;355:171–208. [PubMed: 8781583]
  • Jelmert Ø., Hansteen I.-L., Langård S. Enhanced cytogenetic detection of previous in vivo exposure to mutagens in human lymphocytes after treatment with inhibitors of DNA synthesis and DNA repair in vitro. Mutat. Res. 1992;271:289–298. [PubMed: 1378201]
  • Jiang J., Jordan S.J., Barr D.P., Gunther M.R., Maeda H., Mason R.P. In vivo production of nitric oxide in rats after administration of hydroxyurea. Mol. Pharmacol. 1997;52:1081–1086. [PubMed: 9415718]
  • Johns D.G., Gao W.Y. Selective depletion of DNA precursors: An evolving strategy for potentiation of dideoxynucleoside activity against immunodeficiency virus. Biochem. Pharmacol. 1998;55:1551–1556. [PubMed: 9633990]
  • Kaplan M.E., Mack K., Goldberg J.D., Donovan P.B., Berk P.D., Wasserman L.R. Long-term management of polycythemia vera with hydroxyurea: A progress report. Semin. Hematol. 1986;23:167–171. [PubMed: 3749925]
  • Karon M., Benedict W.F. Chromatid breakage: Differential effect of inhibitors of DNA synthesis during G2 phase. Science. 1972;178:62. [PubMed: 4116011]
  • Khera K.S. A teratogenicity study on hydroxyurea and diphenylhydantoin in cats. Teratology. 1979;20:447–452. [PubMed: 542896]
  • Kihlman B.A., Andersson H.C. Synergistic enhancement of the frequency of chromatid aberrations in cultured human lymphocytes by combinations of inhibitors of DNA repair. Mutat. Res. 1985;150:313–325. [PubMed: 3923342]
  • Kunze E., Graewe T., Scherber S., Weber J., Gellhar P. Cell cycle dependence of Nmethyl-N-nitrosourea-induced tumour development in the proliferating, partially resected urinary bladder. Br. J. exp. Pathol. 1989;70:125–142. [PMC free article: PMC2040540] [PubMed: 2730838]
  • Lambert B., Sten M., Söderhäll S., Ringborg U., Lewensohn R. DNA repair replication, DNA breaks and sister-chromatid exchange in human cells treated with adriamycin in vitro. Mutat. Res. 1983;111:171–184. [PubMed: 6633548]
  • Landaw S.A. Acute leukemia in polycythemia vera. Semin. Hematol. 1986;23:156–165. [PubMed: 3704667]
  • Levin V.A., Prados M.D. Treatment of recurrent gliomas and metastatic brain tumors with a polydrug protocol designed to combat nitrosourea resistance. J. clin. Oncol. 1992;10:766–771. [PubMed: 1314890]
  • Li J.C., Kaminskas E. Progressive formation of DNA lesions in cultured Ehrlich ascites tumor cells treated with hydroxyurea. Cancer Res. 1987;47:2755–2758. [PubMed: 3552205]
  • Liozon E., Brigaudeau C., Trimoreau F., Desangles F., Fermeaux V., Praloran V., Bordessoule D. Is treatment with hydroxyurea leukemogenic in patients with essential thrombocythemia? An analysis of three new cases of leukaemic transformation and review of the literature. Hematol. Cell Ther. 1997;39:11–18. [PubMed: 9088933]
  • Lisziewicz J., Jessen H., Finzi D., Siliciano R.F., Lori F. HIV-1 suppression by early treatment with hydroxyurea, didanosine, and a protease inhibitor. Lancet. 1998;352:199–200. [PubMed: 9683211]
  • Löfvenberg E., Wahlin A. Management of polycythaemia vera, essential thrombocythaemia and myelofibrosis with hydroxyurea. Eur. J. Haematol. 1988;41:375–381. [PubMed: 3197824]
  • Löfvenberg E., Nordenson I., Wahlin A. Cytogenetic abnormalities and leukemic transformation in hydroxyurea-treated patients with Philadelphia chromosome negative chronic myeloproliferative disease. Cancer Genet. Cytogenet. 1990;49:57–67. [PubMed: 2397474]
  • Lori F., Gallo R.C., Malykh A., Cara A., Romano J., Markham P., Franchini G. Didanosine but not high doses of hydroxyurea rescue pigtail macaque from a lethal dose of SIV (smmpbj14). AIDS Res. hum. Retroviruses. 1997a;13:1083–1088. [PubMed: 9282812]
  • Lori F., Malykh A.G., Foli A., Maserati R., De Antoni A., Minoli L., Padrini D., Degli Antoni A., Barchi E., Jessen H., Wainberg M.A., Gallo R.C., Lisziewicz J. Combination of a drug targeting the cell with a drug targeting the virus controls human immunodeficiency virus type 1 resistance. AIDS Res. hum. Retroviruses. 1997b;13:1403–1409. [PubMed: 9359660]
  • Loukopoulos D., Voskaridou E., Stamoulakatou A., Papassotiriou Y., Kalotychou V., Loutradi A., Cozma G., Tsiarta H., Pavlides N. Hydroxyurea therapy in thalassemia. Ann. N.Y. Acad. Sci. 1998;850:120–128. [PubMed: 9668534]
  • Luzzati R., Di-Perri G., Fendt D., Ramarli D., Broccali G., Concia E. Pharmacokinetics, safety and anti-human immunodeficiency virus (HIV) activity of hydroxyurea in combination with didanosine (Letter). J. antimicrob. Chemother. 1998;42:565–566. [PubMed: 9818769]
  • Maier-Redelsperger M., de Montalembert M., Flahault A., Neonato M.G., Ducrocq R., Masson M.P., Girot R., Elion J. for the French Study Group on Sickle Cell Disease, author. Fetal hemoglobin and F-cell responses to long-term hydroxyurea treatment in young sickle cell patients. Blood. 1998;91:4472–4479. [PubMed: 9616141]
  • Mattano S.S., Palella T.D., Mitchell B.S. Mutations induced at the hypoxanthine–guanine phosphoribosyltransferase locus of human T-lymphoblasts by perturbations of purine deoxyribonucleoside triphosphate pools. Cancer Res. 1990;50:4566–4571. [PubMed: 2369732]
  • Mayer V.W., Goin C.J., Zimmermann F.K. Aneuploidy and other genetic effects induced by hydroxyurea in Saccharomyces cerevisiae. Mutat. Res. 1986;160:19–26. [PubMed: 3512984]
  • Medical Economics Data Production (1999) PDR®: Physicians’ Desk Reference, 53rd Ed., Montvale, NJ, Medical Economics.
  • Mehnert K., Vogel W., Benz R., Speit G. Different effects of mutagens on sister chromatid exchange induction in three Chinese hamster cell lines. Environ. Mutag. 1984;6:573–583. [PubMed: 6088219]
  • Millicovsky G., DeSesso J.M., Kleinman L.I., Clark K.E. Effects of hydroxyurea on hemodynamics of pregnant rabbits: A maternally mediated mechanism of embryotoxicity. Am. J. Obstet. Gynecol. 1981;140:747–752. [PubMed: 7196156]
  • Minford J., Kerrigan D., Nichols M., Shackney S., Zwelling L.A. Enhancement of the DNA breakage and cytotoxic effects of intercalating agents by treatment with sublethal doses of 1-beta-D-arabinofuranosylcytosine or hydroxyurea in L1210 cells. Cancer Res. 1984;44:5583–5593. [PubMed: 6208999]
  • de Montalembert M., Belloy M., Bernaudin F., Gouraud F., Capdeville R., Mardini R., Philippe N., Jais J.P., Bardakdjian J., Ducrocq R., Maier-Redelsperger M., Elion J., Labie D., Girot R. for the French Study Group on Sickle Cell Disease, author. Three-year follow-up of hydroxyurea treatment in severely ill children with sickle cell disease. J. pediatr. Hematol./Oncol. 1997;19:313–318. [PubMed: 9256830]
  • Montaner J.S., Zala C., Conway B., Raboud J., Patenaude P., Rae S., O’Shaughnessy M.V., Schechter M.T. A pilot study of hydroxyurea among patients with advanced human immunodeficiency virus (HIV) disease receiving chronic didanosine therapy: Canadian HIV trials network protocol 080. J. infect. Dis. 1997;175:801–806. [PubMed: 9086133]
  • Moschella S.L., Greenwald M.A. Psoriasis with hydroxyurea. An 18-month study of 60 patients. Arch. Dermatol. 1973;107:363–368. [PubMed: 4692124]
  • Muranyi-Kovacs I., Rudali G. Comparative study of carcinogenic activity of hydroxyurea and urethane in XVII-G mice. Rev. Eur. Etud. clin. biol. 1972;17:93–95. [PubMed: 5041353]
  • Murphy M.L., Chaube S. Preliminary survey of hydroxyurea (NSC-32065) as a teratogen. Cancer Chemother. Rep. 1964;40:1–7. [PubMed: 14206908]
  • Nair R., Iyer R.S., Nair C.N., Kurkure P.A., Pai S.K., Saikia T.K., Nadkarni K.S., Pai V.R., Gopal R., Advani S.H. Myelodysplastic syndrome. A clinical and pathological analysis of 88 patients. Indian J. Cancer. 1993;30:169–175. [PubMed: 8206499]
  • Najean Y., Rain J.-D. Treatment of polycythemia vera: The use of hydroxyurea and pipobroman in 292 patients under the age of 65 years. Blood. 1997a;90:3370–3377. [PubMed: 9345019]
  • Najean Y., Rain J.D. for the French Polycythemia Study Group, author. Treatment of polycythemia vera: Use of 32P alone or in combination with maintenance therapy using hydroxyurea in 461 patients greater than 65 years of age. Blood. 1997b;89:2319–2327. [PubMed: 9116275]
  • Najean Y., Rain J.-D., Dresch C., Goguel A., Lejeune F., Echard M., Grange M.-J. Risk of leukaemia, carcinoma, and myelofibrosis in 32P- or chemotherapy-treated patients with polycythemia vera: A prospective analysis of 682 cases. Leuk. Lymphoma. 1996;22:111–119. [PubMed: 8951781]
  • Nand S., Messmore H., Fisher S.G., Bird M.L., Schulz W., Fisher R.I. Leukemic transformation in polycythemia vera: Analysis of risk factors. Am. J. Hematol. 1990;34:32–36. [PubMed: 2327402]
  • Nand S., Stock W., Godwin J., Fisher S.G. Leukemogenic risk of hydroxyurea therapy in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Am. J. Hematol. 1996;52:42–46. [PubMed: 8638610]
  • Navarra P., Del Carmine R., Ciabattoni G., D’Amato M., Ragazzoni E., Vacca M., Volpe A.R., Preziosi P. Hydroxyurea: Relationship between toxicity and centrally-induced adrenal activation. Pharmacol. Toxicol. 1990;67:209–215. [PubMed: 2255677]
  • Navarra P., Puccetti P., Riccardi C., Preziosi P. Anticancer drug toxicity via cytokine production: The hydroxyurea paradigm. Toxicol. Lett. 1995;82–83:167–171. [PubMed: 8597047]
  • Navarra P., Tringali G., Preziosi P. The effects of inhibitors of cyclo-oxygenase, lipoxygenase and nitric oxide synthase pathways on the toxicity of hydroxyurea in adrenalectomized rats. Toxicol. Lett. 1996;86:13–18. [PubMed: 8685915]
  • Navarra P., Grohmann U., Nocentini G., Tringali G., Puccetti P., Riccardi C., Preziosi P. Hydroxyurea induces the gene expression and synthesis of proinflammatory cytokines in vivo. J. Pharmacol. exp. Ther. 1997;280:477–482. [PubMed: 8996231]
  • Navarra P., Tringali G., Preziosi P. Hydroxyurea influences adrenocortical function in humans. Eur. J. clin. Pharmacol. 1998;54:491–492. [PubMed: 9776442]
  • Nyce J., Liu L., Jones P.A. Variable effects of DNA-synthesis inhibitors upon DNA methylation in mammalian cells. Nucleic Acids Res. 1986;14:4353–4367. [PMC free article: PMC339866] [PubMed: 3086840]
  • Ören H., Düzovali O., Yüksel E., Sakizli M., Irken G. Development of acute promyelocytic leukemia with isochromosome 17q after BCR/ABL positive chronic myeloid leukemia. Cancer Genet. Cytogenet. 1999;109:141–143. [PubMed: 10087949]
  • Painter R.B., Cleaver J.E. Radiobiology. Repair replication in HeLa cells after large doses of X-irradiation. Nature. 1967;216:369–370. [PubMed: 6053811]
  • Pajor A., Zimonyi I., Koos R., Lehoczky D., Ambrus C. Pregnancies and offspring in survivors of acute lymphoid leukemia and lymphoma. Eur. J. Obstet. Gynecol. reprod. Biol. 1991;40:1–5. [PubMed: 1855603]
  • Palitti F., Tanzarella C., Degrassi F., De Salvia R., Fiore M., Natarajan A.T. Formation of chromatid-type aberrations in G2 stage of the cell cycle. Mutat. Res. 1983;110:343–350. [PubMed: 6877259]
  • Palitti F., Tanzarella C., Degrassi F., De Salvia R., Fiore M. Enhancement of induced sister chromatid exchange and chromosomal aberrations by inhibitors of DNA repair processes. Toxicol. Pathol. 1984a;12:269–273. [PubMed: 6440266]
  • Palitti F., Degrassi F., De Salvia R., Tanzarella C., Fiore M. Potentiation of induced sister chromatid exchanges and chromatid-type aberrations by inhibitors of DNA synthesis and repair in G2. Basic Life Sci. 1984b;29A:313–318. [PubMed: 6442570]
  • Patel M., Dukes I.A.F., Hull J.D. Use of hydroxyurea in chronic myeloid leukemia during pregnancy: A case report. Am. J. Obstet. Gynecol. 1991;165:565–566. [PubMed: 1892181]
  • Philips F.S., Sternberg S.S. Tests for tumour induction by antitumor agents. Recent Results Cancer Res. 1975;52:29–35. [PubMed: 827794]
  • Popescu N.C., Turnbull D., DiPaolo J.A. Sister chromatid exchange and chromosome aberration analysis with the use of several carcinogens and noncarcinogens. J. natl Cancer Inst. 1977;59:289–293. [PubMed: 406414]
  • Pouchert, C.J. (1981) The Aldrich Library of Infrared Spectra, 3rd Ed., Milwaukee, WI, Aldrich Chemical Co., p. 475.
  • Pouchert, C.J. (1983) The Aldrich Library of NMR Spectra, 2nd Ed., Milwaukee, WI, Aldrich Chemical Co., Vol. 1, p. 671.
  • Pouchert, C.J. (1985) The Aldrich Library of FT-IR Spectra, Milwaukee, WI, Aldrich Chemical Co., Vol. 1, p. 801.
  • Prempree T., Merz T. Does hydroxyurea inhibit chromosome repair in cultured human lymphocytes? Nature. 1969;224:603–604. [PubMed: 5346601]
  • Price C.J., Tyl R.W., Marks T.A., Taschke L.L., Ledoux T.A., Reel J.R. Teratologic and postnatal evaluation of aniline hydrochloride in the Fischer 344 rat. Toxicol. appl. Pharmacol. 1985;77:465–478. [PubMed: 3975914]
  • Quesnel B., Kantarjian H., Pedersen Bjergaard J., Brault P., Estey E., Lai J.L., Tilly H., Stoppa A.M., Archimbaud E., Harousseau J.L., Bauters F., Fenaux P. Therapy-related acute myeloid leukemia with t(8;21), inv(16), and t(8;16): A report on 25 cases and review of the literature. J. clin. Oncol. 1993;11:2370–2379. [PubMed: 8246025]
  • Reiffers J., Dachary D., David B., Bernard P., Marit G., Boisseau M., Brousted A. Megakaryoblastic transformation of primary thrombocythemia. Acta haematol. 1985;73:228–231. [PubMed: 3933245]
  • Richard M., Truchetet F., Friedel J., Leclech C., Heid E. Skin lesions simulating chronic dermatomyositis during long-term hydroxyurea therapy. J. Am. Acad. Dermatol. 1989;21:797–799. [PubMed: 2808796]
  • Rodriguez G.I., Kuhn J.G., Weiss G.R., Hilsenbeck S.G., Eckardt J.R., Thurman A., Rinaldi D.A., Hodges S., Von Hoff D.D., Rowinsky E.K. A bioavailability and pharmacokinetic study of oral and intravenous hydroxyurea. Blood. 1998;91:1533–1541. [PubMed: 9473217]
  • Rossberger S., Andrae U. DNA repair synthesis induced by N-hydroxyurea, acetohydroxamic acid, and N-hydroxyurethane in primary rat hepatocyte cultures: Comparative evaluation using the autoradiographic and the bromodeoxyuridine density-shift method. Mutat. Res. 1985;145:201–207. [PubMed: 3982435]
  • Rote Liste Sekretariat (1998) Rote Liste 1998, Frankfurt, Rote Liste Service GmbH, pp. 86–106, 86–115.
  • Royal Pharmaceutical Society of Great Britain (1999) Martindale, The Extra Pharmacopoeia, 13th Ed., London, The Pharmaceutical Press [MicroMedex Online: Health Care Series]
  • Rutschmann O.T., Opravil M., Iten A., Malinverni R., Vernazza P.L., Bucher H.C., Bernasconi E., Sudre P., Leduc D., Yerly S., Perrin L.H., Hirschel B. for the Swiss HIV Cohort Study, author. A placebo-controlled trial of didanosine plus stavudine, with and without hydroxyurea, for HIV infection. AIDS. 1998;12:F71–F77. [PubMed: 9631134]
  • Sadtler Research Laboratories (1995) Sadtler Standard Spectra, 1981–1995 Supplementary Index, Philadelphia, PA, p. 1.
  • Sato K., Akaike T., Sawa T., Miyamoto Y., Suga M., Ando M., Maeda H. Nitric oxide generation from hydroxyurea via copper-catalyzed peroxidation and implications for pharmacological actions of hydroxyurea. Jpn. J. Cancer Res. 1997;88:1199–1204. [PMC free article: PMC5921347] [PubMed: 9473738]
  • Schrell U.M., Rittig M.G., Anders M., Koch U.H., Marschalek R., Kiesewetter F., Fahlbusch R. Hydroxyurea for treatment of unresectable and recurrent meningiomas. II. Decrease in the size of meningiomas in patients treated with hydroxyurea. J. Neurosurg. 1997;86:840–844. [PubMed: 9126900]
  • Scott W.J., Ritter E.J., Wilson J.G. DNA synthesis inhibition and cell death associated with hydroxyurea teratogenesis in rat embryos. Dev. Biol. 1971;26:306–315. [PubMed: 5158536]
  • Scott J.P., Hillery C.A., Brown E.R., Misiewicz V., Labotka R.J. Hydroxyurea therapy in children severely affected with sickle cell disease. J. Pediatr. 1996;128:820–828. [PubMed: 8648542]
  • Sharon R., Tatarsky I., Ben-Arieh Y. Treatment of polycythemia vera with hydroxyurea. Cancer. 1986;57:718–720. [PubMed: 3943010]
  • Simchen G., Idar D., Kassir Y. Recombination and hydroxyurea inhibition of DNA synthesis in yeast meiosis. Mol. Gen. Genet. 1976;144:21–27. [PubMed: 772412]
  • Simonelli C., Comar M., Zanussi S., De Paoli P., Tirelli U., Giacca M. No therapeutic advantage from didanosine (ddI) and hydroxyurea versus ddI alone in patients with HIV infection. AIDS. 1997;11:1299–1300. [PubMed: 9256953]
  • Skog S., Heiden T., Eriksson S., Wallström B., Tribukait B. Hydroxyurea-induced cell death in human T lymphoma cells as related to imbalance in DNA/protein cycle and deoxyribonucleotide pools and DNA strand breaks. Anticancer Drugs. 1992;3:379–386. [PubMed: 1421434]
  • Sofuni T., Honma M., Hayashi M., Shimada H., Tanaka N., Wakuri S., Awogi T., Yamamoto K.I., Nishi Y., Nakadate M. Detection of in vitro clastogens and spindle poisons by the mouse lymphoma assay using the microwell method: Interim report of an international collaborative study. Mutagenesis. 1996;11:349–355. [PubMed: 8671759]
  • Stasi R., Cantonetti M., Abruzzese E., Papi M., Didona B., Cavalieri R., Papa G. Multiple skin tumors in long-term treatment with hydroxyurea (Letter to the Editor). Eur. J. Haematol. 1992;48:121–122. [PubMed: 1547876]
  • Stehman F.B. Experience with hydroxyurea as a radiosensitizer in carcinoma of the cervix. Semin. Oncol. 1992;19:48–52. [PubMed: 1641657]
  • Stehman F.B., Bundy B.N., Kucera P.R., Deppe G., Reddy S., O’Connor D.M. Hydroxyurea, 5-fluorouracil infusion, and cisplatin adjunct to radiation therapy in cervical carcinoma: A phase I–II trial of the Gynecologic Oncology Group. Gynecol. Oncol. 1997;66:262–267. [PubMed: 9264574]
  • Sterkers Y., Preudhomme C., Laï J.-L., Demory J.-L., Caulier M.-T., Wattel E., Borderssoule D., Bauters F., Fenaux P. Acute myeloid leukemia and myelodysplastic syndromes following essential thrombocythemia treated with hydroxyurea: High proportion of cases with 17p deletion. Blood. 1998;91:616–622. [PubMed: 9427717]
  • Strauss B., Coyle M., McMahon M., Kato K., Dolyniuk M. DNA synthesis, repair and chromosome breaks in eucaryotic cells. Johns Hopkins med. J. 1972;1:111–124. [PubMed: 4262456]
  • Swiss Pharmaceutical Society, ed. (1999) Index Nominum, International Drug Directory, 16th Ed., Stuttgart, Medpharm Scientific Publishers [MicroMedex Online: Health Care Series]
  • Sylvester R.K., Lobell M., Teresi M.E., Brundage D., Dubowy R. Excretion of hydroxyurea into milk. Cancer. 1987;60:2177–2178. [PubMed: 3481556]
  • Tanaka M., Yamazaki Y., Kondo E., Hattori M., Tsushita K., Utsumi M. Achievement of a complete cytogenetic response with hydroxyurea in a patient with chronic myelogenous leukemia. Leuk. Res. 1997;21:465–468. [PubMed: 9225076]
  • Tefferi A. Is hydroxyurea leukemagenic in essential thrombocythemia? (Letter to the Editor). Blood. 1998;92:1459–1460. discussion 1460–1461. [PubMed: 9694740]
  • Tertian G., Tchernia G., Papiernik E., Elefant E. Hydroxyurea and pregnancy (Letter to the Editor). Am. J. Obstet. Gynecol. 1992;166:1868. [PubMed: 1616001]
  • Theisen C.T., Fradkin R., Wilson J.G. Teratogenicity of hydroxyurea in rhesus monkeys. Teratology. 1973;7:A29.
  • Thomas, J., ed. (1998) Australian Prescription Products Guide, 27th Ed., Victoria, Australian Pharmaceutical Publishing, Vol. 1, p. 1406.
  • Timson J. Hydroxyurea. Mutat. Res. 1975;32:115–132. [PubMed: 765790]
  • Tohda H., Oikawa A. Hypoxanthine enhances hydroxyurea-induced sister-chromatid exchanges in Chinese hamster ovary cells. Mutat. Res. 1990;230:235–240. [PubMed: 2374560]
  • Toraason M., Bohrman J.S., Krieg E., Combes R.D., Willington S.E., Zajac W., Langenbach R. Evaluation of the V79 cell metabolic co-operation assay as a screen in vitro for developmental toxicants. Toxicol. in Vitro. 1992;6:165–174. [PubMed: 20732108]
  • Triadou P., Maier-Redelsperger M., Krishnamoorty R., Deschamps A., Casadevall N., Dunda O., Ducrocq R., Elion J., Girot R., Labie D., Dover G., Cornu P. Fetal haemoglobin variations following hydroxyurea treatment in patients with cyanotic congenital heart disease. Nouv. Rev. Fr. Hematol. 1994;36:367–372. [PubMed: 7534399]
  • Tsutsui T., Barrett J.C., Ts’o P.O.P. Morphological transformation, DNA damage, and chromosomal aberrations induced by a direct DNA perturbation of synchronized Syrian hamster embryo cells. Cancer Res. 1979;39:2356–2365. [PubMed: 445434]
  • US Pharmacopeial Convention (1994) The 1995 US Pharmacopeia, 23rd Rev./The National Formulary, 18th Rev., Rockville, MD, pp. 776–777.
  • US Pharmacopeial Convention (1998) USP Dispensing Information, Vol. I, Drug Information for the Health Care Professional, 18th Ed., Rockville, MD, pp. 1607–1609.
  • Van den Berg C.L., McGill J.R., Kuhn J.G., Walsh J.T., De La Cruz P.S., Davidson K.K., Wahl G.M., Von Hoff D.D. Pharmacokinetics of hydroxyurea in nude mice. Anticancer Drugs. 1994;5:573–578. [PubMed: 7858290]
  • Velez A., Lopez-Rubio F., Moreno J.C. Chronic hydroxyurea-induced dermatomyositis-like eruption with severe dermal elastosis. Clin. exp. Dermatol. 1998;23:94–95. [PubMed: 9692318]
  • Vila J., Nugier F., Bargues G., Vallet T., Peyramond D., Hamedi-Sangsari F., Seigneurin J.M. Absence of viral rebound after treatment of HIV-infected patients with didanosine and hydroxycarbamide. Lancet. 1997;350:635–636. [PubMed: 9288048]
  • Villani P., Maserati R., Regazzi M.B., Giacchino R., Lori F. Pharmacokinetics of hydroxyurea in patients infected with human immunodeficiency virus type I. J. clin. Pharmacol. 1996;36:117–121. [PubMed: 8852387]
  • Vorhees C.V., Butcher R.E., Brunner R.L., Sobotka T.J. A developmental test battery for neurobehavioral toxicity in rats: A preliminary analysis using monosodium glutamate calcium carrageenan, and hydroxyurea. Toxicol. appl. Pharmacol. 1979;50:267–282. [PubMed: 505457]
  • Vorhees C.V., Butcher R.E., Brunner R.L., Wootten V., Sobotka T.J. Developmental toxicity of phychotoxicity of FD and C red dye No. 40 (allura red AC) in rats. Toxicology. 1983;28:207–217. [PubMed: 6636206]
  • Voskaridou E., Kalotychou V., Loukopoulos D. Clinical and laboratory effects of long-term administration of hydroxyurea to patients with sickle-cell/beta-thalassaemia. Br. J. Haematol. 1995;89:479–484. [PubMed: 7537527]
  • Wadler S., Haynes H., Schechner R., Rozenblit A., Wiernik P.H. Phase I trial of high-dose infusional hydroxyurea, high-dose infusional 5-fluorouracil and recombinant interferon-alpha-2a in patients with advanced malignancies. Invest. New Drugs. 1996;13:315–320. [PubMed: 8824349]
  • Wangenheim J., Bolcsfoldi G. Mouse lymphoma L5178Y thymidine kinase locus assay of 50 compounds. Mutagenesis. 1988;3:193–205. [PubMed: 3045481]
  • Warner C.W., Sadler T.W., Shockey J., Smith M.K. A comparison of the in vivo and in vitro response of mammalian embryos to a teratogenic insult. Toxicology. 1983;28:271–282. [PubMed: 6648977]
  • Weinfeld A., Swolin B., Westin J. Acute leukaemia after hydroxyurea therapy in polycythemia vera and allied disorders: Prospective study of efficacy and leukaemogenicity with therapeutic implications. Eur. J. Haematol. 1994;52:134–139. [PubMed: 8168592]
  • Weinlich G., Schuler G., Greil R., Kofler H., Fritsch P. Leg ulcers associated with long-term hydroxyurea therapy. J. Am. Acad. Dermatol. 1998;39:372–374. [PubMed: 9703157]
  • West W.O. Hydroxyurea in the treatment of polycythemia vera: A prospective study of 100 patients over a 20-year period. South. med. J. 1987;80:323–327. [PubMed: 3824016]
  • Wiger R., Hongslo J.K., Evenson D.P., De Angelis P., Schwartze P.E., Holme J.A. Effects of acetaminophen and hydroxyurea on spermatogenesis and sperm chromatin structure in laboratory mice. Reprod. Toxicol. 1995;9:21–33. [PubMed: 8520128]
  • Wilson, J.G. (1974) Teratogenic causation in man and its evaluation in non-human primates. In: Motulsky, A.G. & Lenz, W., eds, Birth Defects. Proceedings of 4th International Conference, Amsterdam, Excerpta Medica, pp. 191–203.
  • Wilson J.G., Scott W.J., Ritter E.J., Fradkin R. Comparative distribution and embryotoxicity of hydroxyurea in pregnant rats and rhesus monkeys. Teratology. 1975;11:169–178. [PubMed: 1154282]
  • Zaborowska D., Swietlinska Z., Zuk J. Induction of mitotic recombination by UV and diepoxybutane and its enhancement by hydroxyurea in Saccharomyces cerevisiae. Mutat. Res. 1983;120:21–26. [PubMed: 6339914]

Phenolphthalein

1. Exposure Data

1.1. Chemical and physical data

1.1.1. Nomenclature
  • Chem. Abstr. Serv. Reg. No.: 77-09-8
  • Chem. Abstr. Name: 3,3-Bis(4-hydroxyphenyl)-1-(3H)-isobenzofuranone
  • IUPAC Systematic Name: Phenolphthalein
  • Synonyms: 3,3-Bis(4-hydroxyphenyl)phthalide; 3,3-bis(para-hydroxyphenyl)-phthalide; α-(para-hydroxyphenyl)-α-(4-oxo-2,5-cyclohexadien-1-ylidine)-ortho-toluic acid
1.1.2. Structural and molecular formulae and relative molecular mass
Image 387a
1.1.3. Chemical and physical properties of the pure substance
  • (a) Description: White or yellowish-white, triclinic crystals, often twinned (Budavari, 1996)
  • (b) Melting-point: 258–262 °C (Budavari, 1996)
  • (c) Spectroscopy data: Infrared (prism, [8113; 1471C]; grating [28037]; FT-IR, [1006B]), ultraviolet [2188] and nuclear magnetic resonance (proton, [14709]; 13C, [4455]) spectral data have been reported (Sadtler Research Laboratories, 1980; Pouchert, 1981, 1985)
  • (d) Solubility: Practically insoluble in water; soluble in diethyl ether, ethanol and dilute solutions of alkali hydroxides; very slightly soluble in chloroform (Budavari, 1996)
  • (e) Dissociation constant: pKa at 25 °C, 9.7 (Budavari, 1996)
1.1.4. Technical products and impurities

Phenolphthalein (white) is available as a 6.5-, 14-, 32.4-, 60-, 65-, 75-, 100-, 120-, 130- and 200-mg tablet, a 60- and 120-mg chewable tablet, a 30-, 65- and 90-mg capsule, a 64.8-mg wafer, a 15-, 50-, 60-, 65-, 66.7- and 200-mg/5 mL and 198 mg/15 mL liquid emulsion and a 117-mg (9%) chocolate square; phenolphthalein (yellow) is available as a 65-, 90-, 95- and 135-mg tablet, an 80-, 90-, 95- and 97.2-mg chewable tablet, a 65- and 130-mg capsule and a 97.2-mg chewing gum. The tablets may also contain aloin, aspartame, bile salts, butylparaben, cascara sagrada, cascara sagrada extract, corn starch, cocoa butter, cocoa paste from cocoa seeds, colourants (D&C Yellow No. 10 aluminium lake, D&C Red No. 28, FD&C Blue No. 1, FD&C Red No. 40, FD&C No. 40 aluminium lake), docusate sodium, dextrates, dibasic calcium phosphate dihydrate, ethyl vanillin, flavours, hydroxypropyl methylcellulose, lactose, leaves of senna, lecithin from soya beans, magnesium stearate, methylene blue, micro-crystalline cellulose, oleoresin capsicum, ox bile extract, polydextrose, polyethylene glycol, polysorbate 80, potassium nitrate, povidone, propylene glycol, sodium carbonate (anhydrous), sodium saccharin, sodium starch glycolate, starch, sucrose, titanium dioxide and triacetin. The capsule may also contain dehydrocholic acid, docusate calcium, ethanol, parabens, povidone and sorbitol. The liquid emulsion may also contain agar, benzoic acid, glycerin, liquid paraffin, mineral oil, sodium cyclamate and sorbic acid (Gennaro, 1995; American Hospital Formulary Service, 1997; Canadian Pharmaceutical Association, 1997; Medical Economics Data Production, 1998; Rote Liste Sekretariat, 1998; Thomas, 1998; US Pharmacopeial Convention, 1998).

In the manufacture of phenolphthalein, a stage is reached in which certain by-products formed in the synthesis have not yet been removed, resulting in a product called yellow phenolphthalein. Compounds isolated from one sample of yellow phenolphthalein were: white phenolphthalein, 93%; fluoran, 0.32%; isophenolphthalein, 0.08%; 2-(4-hydroxybenzoyl)benzoic acid, 0.10%. Yellow phenolphthalein was reported to be 2.5 times more active as a laxative in rhesus monkeys than phenolphthalein (Budavari, 1996).

Trade names for phenolphthalein include Alophen Pills, Ap-La-Day, Bonomint, Brooklax, Caolax N.F., Certolax, Cirulaxia, Confetto Falqui, Darmol, Dilsuave, Easylax, Espotabs, Evac-Q-Tabs, Evac-U-Gen, Evac-U-Lax, Ex-Lax, Feen-A-Mint, Figsen, Fletchers Childrens Laxative, Fructines, Fructosan, Lacto-Purga, Laxative Pills, Laxen Busto, Laxettes, Lax-Pills, Lilo, Medilax, Modane, Musilaks, Neo-Prunex, Novopuren, Phenolax, Phenolphthalein Tablets USP 23, Prifinol, Prulet, Purga, Purganol, Purgante, Pürjen Sahap, Reguletts, Sure Lax and Thalinol (National Toxicology Program, 1999; Royal Pharmaceutical Society of Great Britain, 1999; Swiss Pharmaceutical Society, 1999).

Trade names for phenolphthalein that have been discontinued include Alophen, Bom-Bon, Confetto, Euchessina, Fructine-Vichy, Koprol, Laxatone, Laxogen, Neopurghes, Phthalin, Prunetta, Purgen, Purgestol, Spulmako-lax and Trilax.

Trade names for multi-ingredient preparations of phenolphthalein include Abfuhr-dragees, Agarol, Agoral, Aid-Lax, Alofedina, Alophen, Alsiline, Bicholate, Calcium Docuphen, Caroid, Carter Petites Pilules, Carters, Carters Little Pills, Cholasyn, Colax, Damalax, Dialose Plus, Disolan, Docucal-P, Doulax, Doxidan, Emuliquen Laxante, Evac-Q-Kwik, Ex-Lax Extra Gentle Pills, Ex-Lax Light, Falqui, Fam-Lax, Feen-a-mint Pills, Femilax, Ford Pills, Grains de Vals, Herbalax Forte, Juno Junipah, Kest, Kondremul with Phenolphthalein, Laxa, Laxante Bescansa, Laxante Bescansa Aloico, Laxante Olan, Laxante Salud, Laxarol, Laxo Vian, Le 100 B, Lipograsil, Mackenzies Menthoids, Mahiou, Modane Plus, Mucinum, Nylax, Obstinol, Paragar, Paragol, Petrolagar No. 2, Petrolagar with Phenolphthalein, Phillips Gelcaps, Phillips’ Laxative Gelcaps, Phillips’ Laxcaps, Phytolax, Pildoras Zeninas, Sanicolax, Takata, Thunas Bilettes, Triolax, Unilax, Vencipon, Veracolate and Vesilax (Royal Pharmaceutical Society of Great Britain, 1999).

Trade names for preparations containing phenolphthalein which have been discontinued include Agarbil, Amaro Lassativo, Bilagar, Boldolaxine, Boldolaxine Aloes, Confetti Lassativi, Confetto Complex, Correctol, Crisolax, Dietaid, Dragées 19, Emulsione Lassativa, Flamlax, Lactolaxine, Lax-Lorenz, Laxante Geve, Laxante Richelet, Laxativum, Laxicaps, Medimonth, Ormobyl, Pillole Lassative Aicardi, Pillole Schias, Pluribase, Reolina, Rim and Verecolene Complesso.

1.1.5. Analysis

Several international pharmacopoeias specify colorimetric and liquid chromatographic methods for identifying phenolphthalein; visible absorption spectrophotometry and liquid chromatography are used to assay its purity. Phenolphthalein is identified in pharmaceutical preparations by colorimetry and liquid chromatography; liquid chromatography is used to assay for content (British Pharmacopoeial Commission, 1993; US Pharmacopeial Convention, 1994).

Several methods for the analysis of phenolphthalein in various matrices have been reported, which include spectrophotometric, titrimetric, polarographic and chromatographic methods. The chromatographic methods include paper, gas, thin-layer and high-performance liquid chromatography (Al-Shammary et al., 1991).

1.2. Production

Phenolphthalein can be prepared from a mixture of phenol, phthalic anhydride and sulfuric acid which is heated to 120 °C for 10–12 h. The product is extracted with boiling water, and the residue is dissolved in dilute sodium hydroxide solution, filtered, and precipitated with acid (Gennaro, 1995).

It has been reported that 197 tonnes of phenolphthalein were produced by one US manufacturer in the early 1990s (National Toxicology Program, 1999). Information available in 1999 indicated that phenolphthalein was manufactured and/or formulated in 33 countries (CIS Information Services, 1998; Royal Pharmaceutical Society of Great Britain; 1999; Swiss Pharmaceutical Society, 1999).

1.3. Use

Phenolphthalein is a stimulant laxative which has been used for the treatment of constipation and for bowel evacuation before investigational procedures or surgery. The laxative effect of phenolphthalein was discovered in 1902, and it has been widely used since that time (Mvros et al., 1991). It usually has an effect within 4–8 h after oral administration, generally in tablets or capsules; it is also available as an emulsion with liquid paraffin. It is available without prescription in many countries. The usual oral laxative dose of phenolphthalein (white or yellow) is 30–200 mg daily taken at bedtime for adults and children aged ≥ 12 years (270 mg should not be exceeded); 30–60 mg daily for children aged 6–11 years; and 15–30 mg daily for children aged 2–5 years, given as a single or divided doses. A dose of 260 mg has been used in regimens for bowel evacuation (American Hospital Formulary Service, 1997; Royal Pharmaceutical Society of Great Britain, 1999).

The use of laxatives to relieve constipation and to maintain regularity in bowel habits is common in western cultures. Studies in Australia, the United Kingdom and the USA have found that about 20% of the general population reports regular use of laxatives (Kune, 1993). Two large surveys of the adult population in the USA found that about 10% of adults used some form of laxative at least once a month, that female users outnumbered male users and that the fraction of users increases with age (Everhart et al., 1989; Harari et al., 1989).

Few studies report the prevalence of use of phenolphthalein laxatives. One study of 424 cases of colon cancer and 414 controls in Washington State, USA, aged 30–62, found that 34% of the control subjects reported constipation requiring treatment (use of a laxative, enema or prunes), 2.7% reported ever having used phenolphthalein laxatives and 1.4% reported having used phenolphthalein laxatives at least 350 times in their lifetimes (Jacobs & White, 1998).

In three populations of 268–813 persons who had undergone endoscopy for colon polyps, two in North Carolina and one in California, USA, comprising approximately equal numbers of cases and controls, 0.8–4.4% of the control subjects had used phenolphthalein laxatives at least once per week. The two groups in North Carolina comprised subjects aged 30–89 years, 58% and 53% of whom were female; the group in California comprised subjects aged 50–74 years of whom 34% were female. The mean ages of the three groups were comparable (59–62 years). Among controls, the frequent users of phenolphthalein laxatives represented 5.2–30% of all frequent laxative users. In the two studies in North Carolina, 18% of case subjects and 25% of controls reported ever having used phenolphthalein laxatives, and 10% of cases and 7% of controls had used them at least once a month (Longnecker et al., 1997). In a study of colorectal cancer in Melbourne, Australia (Kune, 1993), 9.7% of the 723 subjects reported ever having used phenolphthalein laxatives.

Phenolphthalein in a 1% alcoholic solution is also used as a visual indicator in titrations of mineral and organic acids and most alkalis. Phenolphthalein-titrated solutions are colourless at pH < 8.5 and pink to deep-red at pH > 9 (Budavari, 1996).

1.4. Occurrence

Phenolphthalein is not known to occur as a natural product. No data on occupational exposure were available to the Working Group.

1.5. Regulations and guidelines

Phenolphthalein is listed in the Austrian, Belgian, British, Chinese, Czech Republic, Hungarian, Italian, Swiss and US pharmacopoeias (Royal Pharmaceutical Society of Great Britain, 1999; Swiss Pharmaceutical Society, 1999).

After the publication in 1996 of the results of studies in rodents indicating that phenolphthalein was carcinogenic and genotoxic in several test systems, with damage (loss) of the p53 tumour suppressor gene (Food & Drug Administration, 1999), many countries moved to restrict over-the-counter sales of phenolphthalein-containing laxatives. Both France and Italy have suspended use of phenolphthalein in prescription and over-the-counter pharmaceutical preparations, and the United Kingdom has changed the status of phenolphthalein from an over-the-counter to prescription agent in pharmaceutical preparations (WHO, 1997; Francesco International, 1998; WHO, 1998). Canada has suspended the sale of all products containing phenolphthalein (Canadian Pharmacists Association, 1999). The German Federal Institute for Drugs and Medical Devices recommended that holders of authorizations to market phenolphthalein-containing laxative products withdraw their products from the market because of the potential toxicological risks. The Japanese Pharmaceutical and Medical Safety Bureau of the Ministry of Health and Welfare issued a statement that laxative products containing phenolphthalein had been voluntarily withdrawn by the manufacturers (WHO, 1998). The Food and Drug Administration (1999) issued a final rule establishing that phenolphthalein is not generally recognized as safe and effective.

2. Studies of Cancer in Humans

Studies of the association between colorectal neoplasia and use of phenolphthalein-containing laxatives are summarized in Table 1.

Table 1. Association between colorectal neoplasia and reported use of phenolphthalein-containing laxatives.

Table 1

Association between colorectal neoplasia and reported use of phenolphthalein-containing laxatives.

2.1. Colon cancer

Kune (1993) analysed data on laxative use reported by 685 subjects with colorectal adenocarcinoma diagnosed in 1980–81 in Melbourne, Australia, and 723 controls frequency matched with cases on age and sex. Laxative use throughout adult life was assessed by interview. The relative risk associated with use of commercially produced laxatives was 1.0 (95% confidence interval [CI], 0.86–1.4). Eighty-seven case subjects (13%) and 70 controls (9.7%) reported having used phenolphthalein-containing laxatives (relative risk, 1.4 [95% CI, 0.96–1.9]).

In a case–control study of the association between colon cancer, constipation and use of phenolphthalein–containing laxatives in Washington State, USA (Jacobs & White, 1998), of 659 potential cases identified, 102 died before being approached and 55 were found to be ineligible. Of the 502 remaining cases, data were obtained from 424. Potentially eligible controls were selected by stratified random sampling of subjects in households identified by random-digit dialling, to approximate the distribution by age, sex and county of residence of the case subjects. Of 549 controls thus identified, data were obtained from 414 subjects. Data on laxative use were obtained by telephone interview, and subjects were also asked to complete a mailed food frequency questionnaire. The reference period was up to two years before diagnosis. Regular use was defined as a total use of more than 90 days. The relative risk for colon cancer associated with up to 349 lifetime uses of phenolphthalein-containing laxatives compared with no regular use was 1.0 (95% CI, 0.3–3.7) after adjustment for fibre as percentage of calories. The relative risk for ≥ 350 lifetime uses was 3.9 (95% CI, 1.5–10). Frequent constipation during the 10 years before the reference date (two years before diagnosis) was associated with an increased risk for colon cancer (4.4; 95% CI, 2.1–8.9). When constipation and commercial laxative use were adjusted for mutually, the association with commercial laxative use was no longer apparent, whereas the association with constipation persisted (2.7; 95% CI, 1.4–5.3). The relative risk associated with use of phenolphthalein-containing laxatives adjusted for constipation was 0.42 (95% CI, 0.10–1.7) for < 350 lifetime uses and 1.4 (95% CI, 0.47–4.3) for > 350 uses. [The Working Group noted the difficulty of excluding possible confounding by indication.]

2.2. Colorectal adenomatous polyps

The association between phenolphthalein-containing laxatives and colorectal adenomatous polyps was investigated in a case–control study in Los Angeles (California, USA) in the period 1991–93 and in two case–control studies in North Carolina (USA) in 1988–90 and 1992–95 (Longnecker et al., 1997). In all three studies, cases and controls were selected from among people undergoing an endoscopic procedure (sigmoidoscopy in Los Angeles, colonoscopy in North Carolina); the cases were those found to have polyps. The main indication for this procedure was screening in the Los Angeles study and bleeding in the North Carolina studies. In the Los Angeles study, data on laxative use were collected by personal interview, and subjects were asked about use of specified agents in the year prior to sigmoidoscopy. The agents specified did not include phenolphthalein-containing laxatives but included ‘other laxative preparations’ as a category. If the subject reported use of laxatives in this category, the specific preparation was recorded. In North Carolina, subjects were asked over the telephone about the brand of laxative they used most often. For all three studies, the responses to questions about the preparation used were reviewed without knowledge of the subject’s case or control status, and laxatives were classified as containing phenolphthalein on the basis of brand. In view of these differences between the studies and differences in the eligibility criteria and matching, the three studies were analysed separately. The relative risk for colorectal polyps associated with use of phenolphthalein-containing laxatives at least once a week was 1.8 (95% CI, 0.5–6.2) in Los Angeles (488 cases, 488 controls), 1.0 (95% CI, 0.4–2.2) in North Carolina in 1988–90 (236 cases, 409 controls) and 1.1 (95% CI, 0.2–5.7) in North Carolina in 1992–95 (142 cases, 169 controls).

[The Working Group noted the low statistical power of these studies to detect associations, resulting from the low prevalence of use of phenolphthalein-containing laxatives.]

3. Studies of Cancer in Experimental Animals

Oral administration

Mouse

Groups of 50 male and 50 female B6C3F1 mice, six to seven weeks of age, were given diets containing phenolphthalein (purity, 99.9%) at a concentration of 3000, 6000 or 12 000 mg/kg for two years, equivalent to 0, 300, 600 or 1200 mg/kg bw in males and 0, 400, 800 or 1500 mg/kg bw in females. Only females treated with the highest dose had a significantly decreased rate of survival when compared with controls. The plasma concentrations of total phenolphthalein were similar at all doses. As shown in Table 2, the incidence of histiocytic sarcoma (principally in the liver but also at other sites) was significantly greater in males and females at the two higher doses than in controls. The incidence of malignant lymphoma (all types) was significantly increased in all groups of treated females, but not in males. The incidence of lymphoma of thymic origin was significantly increased in all groups of exposed females and in males at 6000 ppm. As shown in Table 2, the incidence of benign ovarian sex-cord stromal tumours was significantly increased in treated females; the mean historical incidence of all ovarian luteomas was 0.4% (Dunnick & Hailey, 1996; National Toxicology Program, 1996).

Table 2. Incidences of lesions in mice fed diets containing phenolphthalein.

Table 2

Incidences of lesions in mice fed diets containing phenolphthalein.

Groups of 20 female p53+/− heterozygous mice, 7–10 weeks of age, received diets containing phenolphthalein at a concentration of 0 (control), 200, 375, 750, 3000 or 12 000 mg/kg for 26 weeks, equivalent to average daily doses of phenolphthalein of 0, 43, 84, 174, 689 or 2375 mg/kg bw per day. The two lowest concentrations delivered doses of phenolphthalein that were approximately 0.5–1.5 times the recommended human dose based on a mg/m2 body surface area comparison. The incidence of malignant lymphoma of the thymus was significantly increased in heterozygous p53-deficient female mice given the two higher doses. Atypical thymic hyperplasia, seen in 3/20 animals at 750 mg/kg, 3/20 at 3000 mg/kg and 5/20 at 12 000 ppm, was considered to represent proliferative change preceding lymphoma. The incidence of atypical hyperplasia or malignant lymphoma was increased in animals at 750 ppm. The incidence of malignant lymphomas was significantly increased at the two highest doses (0/19 in controls and 1/20, 0/20, 2/20, 17/20 (p < 0.01) and 14/20 (p < 0.01) at the five doses, respectively). Loss of the p53 wild-type allele was found in 2/2 thymic lymphomas from animals at 750 mg/kg, 13/13 at 200 mg/kg and 6/6 at 12 000 mg/kg (Dunnick et al., 1997).

In a study published as an abstract, p53+/− knock-out mice [age not specified] were given phenolphthalein [purity not specified] for 26 weeks by gavage at a dose of 800 or 2400 mg/kg bw per day [number of treatments per week not specified] or in the diet at 2400 mg/kg bw per day [dietary concentration not specified]. [Details of the control groups were not reported.] The experiment was terminated at 26 weeks. The incidences of thymic lymphomas were 3/15, 4/15 and 12/15 in males and 5/15, 8/15 and 14/15 in females receiving 800 (by gavage), 2400 (by gavage) and 2400 (in the diet) mg/kg bw, respectively (Furst et al., 1999).

Rat

Groups of 50 male and 50 female Fischer 344 rats, seven weeks of age, were given diets containing phenolphthalein (purity, 99.9%) at a concentration of 0, 12 000, 25 000 or 50 000 mg/kg for two years, equivalent to 0, 500, 1000 or 2000 mg/kg bw for males and 0, 500, 1000 or 2500 mg/kg bw for females. As in the mice, the total plasma concentrations of phenolphthalein did not increase with increasing dose. The survival rate in all groups of treated animals was similar to that of controls. As shown in Table 3, the incidence of benign phaeochromocytoma of the adrenal medulla was significantly increased in all treated male groups, and most were bilateral. The incidence of malignant phaeochromocytoma was not increased by treatment at any dose. The incidence of benign phaeochromocytoma was also increased in female rats given the highest dose, but the incidences of bilateral tumours and malignant phaeochromocytoma were not increased in females. As seen in Table 4, the incidence of renal tubular adenoma (single and step sections combined) was also significantly increased in all treated male groups, and a few renal tubular carcinomas were also observed. In females, one renal tubular adenoma was observed at the highest dose (Dunnick & Hailey, 1996; National Toxicology Program, 1996). [The Working Group noted the high doses administered.]

Table 3.. Incidences of lesions in the adrenal medulla in Fischer 344 rats fed diets containing phenolphthalein.

Table 3.

Incidences of lesions in the adrenal medulla in Fischer 344 rats fed diets containing phenolphthalein.

Table 4. Incidences of renal tubular lesions in male Fischer 344 rats fed diets containing phenolphthalein.

Table 4

Incidences of renal tubular lesions in male Fischer 344 rats fed diets containing phenolphthalein.

4. Other Data Relevant to an Evaluation of Carcinogenicity and its Mechanisms

4.1. Absorption, distribution, metabolism and excretion

4.1.1. Humans

The absorption of phenolphthalein in humans has been estimated to be 15% of an oral dose (American Hospital Formulary Service, 1995). The absorbed compound is excreted primarily in the urine as phenolic-hydroxyglucuronide or sulfate conjugates. Some conjugated compound is also excreted in the faeces via the bile, and the resulting enterohepatic recirculation probably contributes to prolongation of the laxative effect (Hardman et al., 1996), a hypothesis supported by the observation that phenolphthalein is ineffective as a laxative in patients suffering from obstructive jaundice or in experimental animals with ligated common bile ducts (Steigmann et al., 1938). Small doses of phenolphthalein (30–60 mg) are excreted by humans entirely as conjugated metabolites in urine or faeces, while larger doses (300 mg) result in excretion of both the free and conjugated drug (Williams, 1959). Use of phenolphthalein by women during breast-feeding may cause diarrhoea in their infants (Tyson et al., 1937).

4.1.2. Experimental systems

Phenolphthalein is absorbed in the intestine (Visek et al., 1956) and is almost completely converted to its glucuronide during extensive first-pass metabolism in the intestinal epithelium and liver (Parker et al., 1980) via uridine diphosphate glucuronosyltransferase (UDPGT) in rodents and dogs (Sund & Hillestad, 1982; National Toxicology Program, 1996). In guinea-pigs, small amounts of sulfate-conjugated metabolites have been detected in isolated mucosal sheets originating in the jejunum and colon (Sund & Lauterbach, 1986). Faecal excretion is the major route of elimination of phenolphthalein in rats, while in mice both urinary and faecal elimination are important. The metabolites identified in urine and faeces are phenolphthalein glucuronide, phenolphthalein sulfate and phenolphthalein hydroxide (Griffin et al., 1998; see Figure 1).

Figure 1

Figure 1

Metabolism of [14C]phenolphthalein in Fischer 344 rats and B6C3F1 mice

Six hours after an intravenous injection of [3H]phenolphthalein to female Wistar rats, analysis of the systemic circulation showed that all of the radiolabel was associated with the glucuronide conjugate (Colburn et al., 1979). Enterohepatic recirculation is limited by the rate of hydrolysis of phenolphthalein glucuronide to aglycone by intestinal bacterial β-glucuronidase (Bergan et al., 1982; National Toxicology Program, 1996).

The extent of enterohepatic recirculation of phenolphthalein was examined in rats with cannulated bile ducts. Within 24 h, 95% of a dose of 25 mg/kg bw [3H]phenolphthalein administered intraperitoneally to female Wistar rats was recovered as glucuronide in the bile, with 0.2% in the urine. In rats without cannulated bile ducts, 86% of the same dose was recovered in the faeces, with little glucuronide, and 10% was recovered in the urine, primarily as the glucuronide (Millburn et al., 1967; Parker et al., 1980).

In male Sprague-Dawley CR-1 strain rats with cannulated femoral veins, femoral arteries and bile ducts given an intravenous dose of 3, 30 or 60 mg phenolphthalein, 99.5% of the dose was eliminated in the bile as the glucuronide. When the same rats were given 3, 30 or 100 mg phenolphthalein glucuronide by intravenous administration, no phenolphthalein was detected in the bile (Mehendale, 1990).

Studies in dogs and mice given [14C]phenolphthalein showed that the radiolabel is evenly distributed throughout the body. In newborn pups of bitches given 4.8 mg/kg bw orally 50 h before whelping, < 0.03% of the dose was found in the liver and gall-bladder and none in the blood, indicating extremely limited passage across the placenta (Visek et al., 1956).

Phenolphthalein is excreted in bile, urine, faeces and milk. In mice, 56% of an oral dose was recovered from the urine within 48 h and an additional 38% from the faeces. When an intravenous dose was given, 30% was recovered from the urine and 68% from the faeces (Visek et al., 1956). Some phenolphthalein is excreted into the bile, and the prolonged cathartic effect may be due to the ensuing enterohepatic recirculation (Hardman et al., 1996). Pre-treatment with hepatic microsomal enzyme inducers increased biliary excretion of metabolites in rats, but post-treatment with enzyme inhibitors decreased it (National Toxicology Program, 1996).

Within 72 h of oral administration of 4.8 mg/kg bw [14C]phenolphthalein to mongrel bitches, 51% of the radiolabel was excreted in the faeces and 36% in the urine. After an intravenous dose, 54% was found in the faeces and 37% in the urine. When the same animals received a cannula in the bile-duct and were given an oral dose, 31% of the radiolabel was found in faeces, 38% in urine and 22% in bile. After an intravenous dose, 11% was eliminated in faeces, 35% in urine and 43% in bile (Visek et al., 1956).

The profile of systemic blood concentration–time for phenolphthalein during 24 h after a single intravenous bolus injection was described by a classical compartmental pharmacokinetics model, with evidence of enterohepatic recirculation (Colburn et al., 1979).

In the two-year bioassays of the National Toxicology Program (1996), the concentrations of total phenolphthalein in plasma were 100–200 µg/mL.

Whole-body autoradiography of male BOM:NMRI mice showed high concentrations of radiolabel in the stomach, gall-bladder and small intestine 1 h after administration of an intragastric dose of 1 mL/100 g bw [14C]phenolphthalein (10 µCi/100g) [10 mL/kg bw or 3.2 mg/kg bw]. As evidenced by the presence of radiolabel in peripheral organs (including the kidney, liver and skin), the compound was absorbed. After 2 h, it had arrived in the large intestine, and 4 h after administration, maximum radiolabel was observed in the rectum. Two days after administration, no radiolabel was detected (Sund et al., 1986).

4.2. Toxic effects

4.2.1. Humans

Until the mid-1990s, phenolphthalein was regarded as non-toxic and safe for consumption, although therapeutic oral doses occasionally produced abdominal discomfort, diarrhoea, nausea, decreased blood pressure and faintness (American Hospital Formulary Service, 1995). Serious side-effects were reported in cases of habitual phenolphthalein consumption under conditions of abuse (Cooke, 1977; Pietrusko, 1977).

The main target organ for the toxic effects of phenolphthalein is reported to be the intestine. Indiscriminate use of phenolphthalein results in chronic constipation and laxative dependence, loss of normal bowel function and bowel irritation. Habitual use for several years may cause a ‘cathartic colon’, i.e. a poorly functioning colon with atonic dilatation, especially on the right side, resulting in extensive retention of the bowel contents. The clinical condition, which resembles chronic ulcerative colitis both radiologically and pathologically, involves thinning of the intestinal wall and loss of the normal mucosal pattern of the terminal ileum (Cummings, 1974; Cummings et al., 1974; Cooke, 1977; Pietrusko, 1977; American Hospital Formulary Service, 1995).

Anecdotal cases of long-term use or overdose of phenolphthalein have been associated with abdominal pain, diarrhoea, vomiting, electrolyte imbalance (hypokalaemia, hypocalcaemia and/or metabolic acidosis or alkalosis), dehydration, malabsorption, protein-losing gastroenteropathy, steatorrhoea, anorexia, weight loss, polydipsia, polyuria, cardiac arrhythmia, muscle weakness, prostration and histopathological lesions (Heizer et al., 1968; Velentzas & Ikkos, 1971; Cummings, 1974; LaRusso & McGill, 1975; Pohl & Lowe, 1978; American Hospital Formulary Service, 1995). Kidney, muscle and central nervous system disturbances are thought to be due to electrolyte imbalance. Loss of intestinal sodium and water stimulates compensatory renin production and secondary aldosteronism, leading to sodium conservation and potassium loss by the kidney. The hypokalaemia contributes to renal insufficiency and is sometimes associated with rhabdomyolysis (Copeland, 1994).

Abuse of phenolphthalein-containing laxatives has been associated with gastrointestinal bleeding, iron-deficient anaemia (Weiss & Wood, 1982), acute pancreatitis (Lambrianides & Rosin, 1984) and multiple organ damage in cases of massive overdose, including fulminant hepatic failure and disseminated intravascular coagulation (Sidhu et al., 1989).

Allergy to phenolphthalein is often manifested as cutaneous inflammatory reactions or fixed drug eruptions, i.e. solitary or multiple, well-defined, erythematous macules that may progress to vesicles and/or bullae. These lesions characteristically recur in the same location with each subsequent dose of phenolphthalein and generally leave residual hyperpigmentation that increases in intensity with each exposure; numerous melanin-containing dermal macrophages have been found in pigmented areas (Wyatt et al., 1972; Davies, 1985; Stroud & Rosio, 1987; Zanolli et al., 1993). In extreme cases, recurrences have involved progressively more severe lesions characterized as bullous erythema multiforme, with focal haemorrhage and necrosis and perivascular lymphocytic infiltration (Shelley et al., 1972) and, in one case report, toxic epidermal necrolysis (Kar et al., 1986).

A review of 204 cases of phenolphthalein ingestion in children aged five years and younger reported to the Pittsburgh Poison Center (USA) over a 30-month period indicated that ingestion of ≤ 1 g was associated with a minimal risk of developing dehydration due to excessive diarrhoea and resulting fluid loss (Mrvos et al., 1991). Despite the profile of low acute toxicity documented in this study, cases of fatal poisoning of children have been reported; symptoms of pulmonary and cerebral oedema, multiple organ effects and encephalitis were attributed to hypersensitivity reactions (Cleves, 1932; Kendall, 1954; Sarcinelli et al., 1970). Repeated administration of phenolphthalein-containing laxatives to children has led to serious illness and multiple hospitalizations (Sugar et al., 1991; Ayass et al., 1993).

4.2.2. Experimental systems

Fischer 344/N rats and B6C3F1 mice were given an NIH 07 diet containing phenolphthalein at a concentration of 0, 3000, 6000, 12 000, 25 000 or 50 000 mg/kg ad libitum for 13 weeks, equivalent to intakes of 0, 200, 400, 800, 1600 or 3500 mg/kg bw for rats, 500, 1000, 2000, 4100 or 9000 mg/kg bw for male mice and 600, 1200, 2400, 5000 or 10 500 mg/kg bw for female mice. Phenolphthalein did not appear to be toxic in rats, and no laxative effect was observed. Rats at the two higher doses showed slightly lower weight gain. Treated rats showed increased relative (to body weight) kidney weights (males only) and elevated absolute and relative liver weights at concentrations of 12 000–50 000 ppm. Female rats showed no effect on body-weight gain, but those receiving concentrations of 6000–50 000 mg/kg had elevated liver weights. The primary treatment-related findings in mice involved the reproductive and haematopoietic systems. The haematopoietic changes included bone-marrow hypoplasia (at 12 000–50 000 mg/kg) and increased splenic haematopoiesis (males only; 25 000 and 50 000 mg/kg) (National Toxicology Program, 1996).

In female mice [strain not specified] fed 5, 25 or 50 mg/kg bw phenolphthalein per day orally for 135 days, no toxic manifestations or evidence of histopathological changes were found in the liver, kidney or gastrointestinal tract (Visek et al., 1956).

Phenolphthalein at doses of 25 and 50 µg/mL was cytotoxic in cultured Chang liver cells, causing decreased cell growth and increased anaerobic glycolysis, i.e. increased glucose consumption and lactate production (Nishikawa, 1981).

4.3. Reproductive and prenatal effects

4.3.1. Humans

No data were available to the Working Group.

4.3.2. Experimental systems

Phenolphthalein is a partial oestrogen in immature rat uteri. Doses of 1–10 mg given subcutaneously twice daily for two days to female Wistar rats weighing 35–40 g induced a dose-related increase in uterine weight, but the maximum increase was only about half of that induced by oestradiol. Phenolphthalein was shown to bind to the oestrogen receptor and was a competitive antagonist to oestradiol (Nieto et al., 1990).

In a study reported in an abstract, exposure of female B6C3F1 mice to 1895 mg/kg bw phenolphthalein orally [method not stated] daily for 30 or 60 days caused no changes in weight gain, oestrous cycles or the numbers of oocyte-containing follicles of any class (primordial, primary, growing or antral), or any detectable pathological change in ovarian cells (Hoyer et al., 1997).

Using a continuous breeding protocol, Chapin et al. (1997a) administered phenolphthalein in the feed of Swiss CD-1 mice at a concentration of 0.1, 0.7 or 3.0% w/v, to provide estimated intakes of 0.15, 1.0 and 4.5 g/kg bw per day (National Toxicology Program, 1996; Chapin et al., 1997b). Pairs of 40 control and 20 treated mice were housed together and allowed to produce up to five litters, the last of which was reared and their reproductive performance measured. Significant reproductive toxicity was observed at the intermediate and high doses. At the intermediate dose, the proportions of pairs producing one to five litters were 100, 89, 84, 68 and 36%, the percentages producing second to fifth litters being significantly smaller than in controls. The decrease at the high dose was more severe, only 5% of pairs producing a fifth litter. Overall, the mean number of litters per pair was reduced by 24 and 50% at the intermediate and high doses, and the number of pups per litter decreased by 58–59%. The final litters were reared on the same diets as the parents. Up to 70% of the pups at the high dose died within four days of birth. Cross-over breeding of animals at the intermediate dose with controls showed that the fertility of the females was affected, the litter sizes being reduced to half. Breeding of the F1 offspring at the intermediate dose with controls showed that treatment halved the number of litters and the litter size. The survival of the F2 pups was not affected. Examination of F0 males at the intermediate dose showed a reduction in testis weight by 36% and in the epididymal sperm count by 30%, and seminiferous tubular degeneration was seen in 9 of 10 treated males. The oestrous cycles and ovarian histology of females at this dose were not affected. Very similar results were found in the F1 adults at termination. No adverse effects were observed at the low dose.

After 13 weeks of exposure to the same doses as used in the studies of toxicity, there was no evidence of reproductive toxicity in female B6C3F1 mice or male or female Fischer 344/N rats. Lower epididymal weights and lower sperm density (number of sperm/g of crude epididymal tissue) were observed in male mice at 12 000, 25 000 and 50 000 mg/kg (National Toxicology Program, 1996).

4.4. Genetic and related effects

4.4.1. Humans

No data were available to the Working Group.

4.4.2. Experimental systems

The results of these studies are summarized in Table 5.

Table 5. Genetic and related effects of phenolphthalein.

Table 5

Genetic and related effects of phenolphthalein.

Phenolphthalein was not mutagenic in several assays in Salmonella typhimurium strains TA1535, TA1537, TA1538, TA98 and TA100 in the presence or absence of exogenous metabolic activation. It did not induce DNA damage in DNA repair-deficient strains of Bacillus subtilis.

Phenolphthalein did not induce sister chromatid exchange in Chinese hamster ovary cells in the presence or absence of exogenous metabolic activation, but it induced a dose-related response in chromosomal aberrations in these cells only in the presence of exogenous metabolic activation.

In experiments in which a number of end-points were studied in Syrian hamster embryo cells (a mixed population of cell types that retain some endogenous metabolizing enzymic activity, including oxidation and peroxidation), phenolphthalein induced chromosomal aberrations and Hprt mutations, but not ouabain mutations or aneuploidy. No evidence was found for adduct formation in DNA of these cells. The data for micronuclei failed to reach statistical significance (p = 0.057). Phenolphthalein caused cellular transformation in the same cell line, indicating that it is metabolized appropriately in this system.

Phenolphthalein increased the incidence of micronucleated erythrocytes in male and female B6C3F1 mice and in male Swiss CD-1 mice. [The Working Group noted that the doses were significantly higher than those to which humans would be exposed.]

Tice et al. (1998) studied the effects of phenolphthalein at various concentrations in the diet of transgenic female mice heterozygous for the p53 gene, over a six-month period. They found significant increases in the frequency of micronucleated erythrocytes, most of which appeared to arise from whole chromosomes rather than chromosomal damage; these were observed at doses comparable to those to which humans are exposed. Inconclusive evidence was found for DNA damage in blood leukocytes, and there was no evidence for DNA damage, apoptosis or necrosis in liver parenchymal cells.

In phenolphthalein-induced thymic lymphomas in B6C3F1 mice, p53 protein accumulated in most tumour cell nuclei, but detectable p53 protein was not seen in control thymuses in this model (Dunnick et al., 1997). Other studies have shown that accumulation of p53 protein results from p53 gene alterations (Hegi et al., 1993).

In p53+/− heterozygous mice, phenolphthalein induced atypical hyperplasia and malignant lymphomas of thymic origin within six months in 0% of controls, 5% of animals at 200 mg/kg, 5% at 375 mg/kg, 25% at 750 mg/kg, 100% at 3000 mg/kg and 95% of animals at 12 000 mg/kg. Two of two thymic lymphomas examined from animals at 750 mg/kg, 13/13 from those at 3000 mg/kg and 6/6 from those at 12 000 mg/kg had lost the remaining p53 wild-type allele (Dunnick et al., 1997). No spontaneous thymic lymphomas were found in control mice in these studies, but in other studies in p53+/− mice of spontaneous tumours (which may occur in mice after one year of age), only 55% showed loss of the remaining functional p53 allele (Harvey et al., 1993). The presence of functional p53 protein is essential for normal cell growth. When this protein is absent, as is the case in phenolphthalein-induced thymic lymphomas, regulation of cell cycle electrophoresis is lost and malignant progression may be enhanced.

4.5. Mechanistic considerations

Analogy with related biphenolic compounds suggests that phenolphthalein has oestrogenic activity; however, studies with MCF-7 human breast cancer cells in tissue culture (Ravdin et al., 1987) and in rat uterus in vivo (Nieto et al., 1990) suggested only a weak oestrogenic response. Tsutsui et al. (1997) used the nuclease P1 enhancement version of the 32P-postlabelling assay to investigate whether (and what type of) DNA adducts were responsible for the morphological transformation induced by phenolphthalein. Although they found no adducts, they recognized the possible limitations of the techniques and suggested that small DNA adducts formed by free radicals could be involved in the effects. Sipe et al. (1997) showed free radical metabolism of phenolphthalein by peroxidases in vitro.

The observation of Witt et al. (1995) that chromosomal damage in Chinese hamster ovary cells occurred only when exogenous metabolic activation was added suggests that some as yet unidentified metabolite is responsible for these effects. Bishop et al. (1998) also interpreted differences in the micronucleus response between the two human lymphoblastoid cell lines, MCL-5 and AHH-1 TK+/−, as being likely to reflect the importance of a metabolite in chromosome-damaging effects.

Tice et al. (1998) suggested that numerical chromosomal loss is responsible for the enhanced incidence of thymic tumours seen after treatment with phenolphthalein in p53 heterozygous mice. Dunnick et al. (1997) noted that these tumours uniformly showed loss of heterozygosity for the p53 allele rather than point mutations, suggesting either chromosome loss or deletions of large chromosomal segments.

The ability to detect micronuclei but not mutations at the TK locus in AHH cells may indicate that cells containing phenolphthalein-induced lesions are susceptible to apoptosis (Bishop et al., 1998).

5. Summary of Data Reported and Evaluation

5.1. Exposure data

Phenolphthalein has been widely used as a laxative for nearly a century. Generally available without prescription, it is now being withdrawn from the market in many countries because of recent toxicological concern. Phenolphthalein has also long been used in the laboratory as an indicator in acid–base titrations.

5.2. Human carcinogenicity data

In the few available studies, there was no consistent association between the occurrence of colon cancer or adenomatous colorectal polyps and use of phenolphthalein-containing laxatives. Cancers at other sites have not been studied.

5.3. Animal carcinogenicity data

Phenolphthalein was tested for carcinogenicity by oral administration in two experiments in mice and in one experiment in rats. In one experiment in mice, it induced histiocytic sarcomas and lymphomas in both males and females and benign ovarian tumours in females. In an experiment in mice lacking one allele of the p53 tumour suppressor gene, it increased the incidence of lymphomas. This result was confirmed in a separate study reported as an abstract. It induced benign renal tumours in male rats and benign phaeochromocytomas in males and females.

5.4. Other relevant data

Phenolphthalein is absorbed in the small bowel and is conjugated in the liver to form phenolphthalein glucuronide, which is eliminated in the bile. As it passes through the small intestine, it is partially deconjugated and reabsorbed.

Phenolphthalein and its glucuronide enhance oxygen radical production and cause oxidative damage in vitro. Phenolphthalein has also been shown to have low oestrogenic activity in some model systems. Phenolphthalein induced micronucleated erythrocytes in mice given multiple but not single treatments by gavage or in feed. Abnormal spermatozoa were induced in male mice but not male rats treated with phenolphthalein in the feed for 13 weeks. The malignant thymic lymphomas induced by phenolphthalein in female heterozygous p53-deficient mice showed loss of the normal p53 allele.

Phenolphthalein induced chromosomal aberrations, Hprt gene mutations and morphological transformation but not aneuploidy or ouabain-resistant mutations or sister chromatid exchange in cultured mammalian cells. It did not induce gene mutations in bacteria.

5.5. Evaluation

There is inadequate evidence in humans for the carcinogenicity of phenolphthalein.

There is sufficient evidence in experimental animals for the carcinogenicity of phenolphthalein.

Overall evaluation

Phenolphthalein is possibly carcinogenic to humans (Group 2B).

6. References

  • Al-Shammary, F., Mian, M.S. & Mian, N.A.A (1991) Phenolphthalein. In: Florey, K., ed., Analytical Profiles of Drug Substances, New York, Academic Press, Vol. 20, pp. 627–664.
  • American Hospital Formulary Service (1995) AHFS Drug Information® 95, Bethesda, MD, American Society of Health-System Pharmacists, pp. 1986–1995.
  • American Hospital Formulary Service (1997) AHFS Drug Information® 97, Bethesda, MD, American Society of Health-System Pharmacists, pp. 2241–2242.
  • Ayass, M., Bussing, R. & Mehta, P. (1993) Munchausen syndrome presenting as hemophilia: A convenient and economical ‘steal’ of disease and treatment. Pediatr. Hematol. Oncol., 10, 241–244. [PubMed: 8217539]
  • Bergan T., Fotland M.H., Sund R.B. Interaction between diphenolic laxatives and intestinal bacteria in vitro. Acta pharmacol. toxicol. 1982;51:165–172. [PubMed: 6896788]
  • Bishop M.E., Aidoo A., Domon O.E., Morris S.M., Casciano D.A. Phenolphthalein induces micronuclei in transgenic human lymphoblastoid cells. Environ. mol. Mutag. 1998;32:286–288. [PubMed: 9814444]
  • Bonin A.M., Farqhuarson J.B., Baker R.S.U. Mutagenicity of arylmethane dyes in Salmonella. Mutat. Res. 1981;89:21–34. [PubMed: 6165887]
  • British Pharmacopoeial Commission (1993) British Pharmacopoeia 1993, London, Her Majesty’s Stationery Office, Vol. I, pp. 502–503.
  • Budavari, S., ed. (1996) The Merck Index, 12th Ed., Whitehouse Station, NJ, Merck & Co., pp. 1248, 1726–1727.
  • Canadian Pharmaceutical Association (1997) CPS Compendium of Pharmaceuticals and Specialties, 32nd Ed., Ottawa, Ontario, pp. 40, 490, 564.
  • Canadian Pharmacists Association (1999) Drug Brief—Phenolphthalein [http://www​.cdnpharm.ca/drugbrf2.htm]
  • Chapin R., Gulati D., Mounce R., Russell S. Phenolphthalein. Environ. Health Perspectives. 1997a;105:335–336.
  • Chapin R.E., Sloane R.A., Hasemen J.K. The relationships among reproductive endpoints in Swiss mice, using the Reproductive Assessment by Continuous Breeding database. Fundam. appl. Toxicol. 1997b;38:129–142. [PubMed: 9299186]
  • CIS Information Services (1998) Worldwide Bulk Drug Users Directory 1997/98 Edition, Dallas, TX [CD-ROM]
  • Cleves M. Poisoning by Exlax tablets. J. Am. med. Assoc. 1932;99:654–655.
  • Colburn W.A., Hirom P.C., Parker R.J., Milburn P. A pharmacokinetic model for enterohepatic recirculation in the rat: Phenolphthalein, a model drug. Drug Metab. Dispos. 1979;7:100–102. [PubMed: 38070]
  • Cooke W.T. Laxative abuse. Clin. Gastroenterol. 1977;6:659–673. [PubMed: 923143]
  • Copeland P.M. Renal failure associated with laxative abuse. Psychother. Psychosom. 1994;62:200–202. [PubMed: 7531354]
  • Cummings J.H. Laxative abuse. Gut. 1974;15:758–766. [PMC free article: PMC1413039] [PubMed: 4435592]
  • Cummings J.H., Sladen G.E., James O.F.W., Sarner M., Misiewicz J.J. Laxative-induced diarrhoea: A continuing clinical problem. Br. med. J. 1974;i:537–541. [PMC free article: PMC1633677] [PubMed: 4817188]
  • Davies, D.M., ed. (1985) Textbook of Adverse Drug Reactions, 3rd Ed., New York, Oxford University Press, pp. 110, 121, 475–479.
  • Dietz D.D., Elwell M.R., Chapin R.E., Shelby M.D., Thompson M.B., Filler R., Stedham M.A. Subchronic (13 week) toxicity studies of phenolphthalein in Fischer 344 rats and B6C3F1 mice. Fundam. appl. Toxicol. 1992;18:48–58. [PubMed: 1601209]
  • Dunnick J.K., Hailey J.R. Phenolphthalein exposure causes multiple carcinogenic effects in experimental model systems. Cancer Res. 1996;56:4922–4926. [PubMed: 8895745]
  • Dunnick J.K., Hardisty J.F., Herbert R.A., Seely J.C., Furedi-Machacek E.M., Foley J.F., Lacks G.D., Stasiewicz S., French J.E. Phenolphthalein induces thymic lymphomas accompanied by loss of the p53 wild type allele in heterozygous p53-deficient (±) mice. Toxicol. Pathol. 1997;25:533–540. [PubMed: 9437796]
  • Everhart J.E., Go V.L.W., Johannes R.S., Fitzsimmons S.C., Roth H.P., White L.R. A longitudinal survey of self-reported bowel habits in the United States. Dig. Dis. Sci. 1989;34:1153–1162. [PubMed: 2787735]
  • Food Drug Administration, author. Laxative drug products for over-the-counter human use. Fed. Regist. 1999;64:4535–4540. [PubMed: 10557608]
  • Francesco International (1998) Major Rx-to-OTC Switched Products: The Important Switches of 1997 in Major Markets [http://www​.rxtootcswitch.com/data.htm]
  • Furst S.M., Blanchard K.T., Lilly P.D., Holden H.E., Stoltz J.H., Barthel C., Stoll R.E. Six month oral gavage and diet carcinogenicity study with phenolphthalein in the heterozygous p53 +/− mouse (Abstract No. 1202). Toxicologist. 1999;48
  • Gennaro, A.R. (1995) Remington: The Science and Practice of Pharmacy, 19th Ed., Easton, PA, Mack Publishing, Vol. II, p. 897.
  • Gibson D.P., Brauninger R., Shaffi H.S., Kerckaert G.A., LeBoeuf R.A., Isfort R.J., Aardema M.J. Induction of micronuclei in Syrian hamster embryo cells: Comparison to results in the SHE cell transformation assay for National Toxicology Program test chemicals. Mutat. Res. 1997;392:61–90. [PubMed: 9269331]
  • Griffin R.J., Godfrey V.B., Burka L.T. Metabolism and disposition of phenolphthalein in male and female F344 rats and B6C3F1 mice. Toxicol. Sci. 1998;42:73–81. [PubMed: 9579019]
  • Harari D., Gurwitz J.H., Avorn J., Bohn R., Minaker K.L. Bowel habit in relation to age and gender. Findings from the National Health Interview Survey and clinical implications. Arch. intern. Med. 1989;156:315–320. [PubMed: 8572842]
  • Hardman, J.G., Limbird, L.E., Molinoff, P.B. & Ruddon, R.W., eds (1996) Goodman and Gilman’s The Pharmacological Basis of Therapeutics, 9th Ed., New York, McGraw-Hill, pp. 921–925.
  • Harvey M., McArthur M.J., Montgomery C.A. Jr, Butel J.S., Bradley A., Donehower L.A. Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice. Nature Genet. 1993;5:225–229. [PubMed: 8275085]
  • Hegi M.E., Söderkvist P., Foley J.F., Schoonhoven R., Swenberg J.A., Kari F., Maronpot R., Anderson M.W., Wiseman R.W. Characterization of p53 mutations in methylene chloride-induced lung tumors from B6C3F1 mice. Carcinogenesis. 1993;14:803–810. [PubMed: 8504472]
  • Heizer W.D., Warshaw A.L., Waldmann T.A., Laster L. Protein-losing gastroenteropathy and malabsorption associated with factitious diarrhea. Ann. intern. Med. 1968;68:839–852. [PubMed: 5642966]
  • Hoyer P.B., Boese B., Sipes I.G. The effect of phenolphthalein on B6C3F1 mouse ovaries (Abstract No. 1825). Toxicologist. 1997;36:359.
  • Jacobs E.J., White E. Constipation, laxative use, and colon cancer among middle-aged adults. Epidemiology. 1998;9:385–391. [PubMed: 9647901]
  • Kada T., Tutikawa K., Sadaie Y. In vitro and host-mediated ‘rec-assay’ procedures for screening chemical mutagens; and phloxine, a mutagenic red dye detected. Mutat. Res. 1972;16:165–174. [PubMed: 4342303]
  • Kar P.K., Dutta R.K., Shah B.H. Toxic epidermal necrolysis in a patient induced by phenolphthalein. J. Indian med. Assoc. 1986;84:189–190, 193. [PubMed: 3559223]
  • Kendall A.C. Fatal case of encephalitis after phenolphthalein ingestion. Br. med. J. 1954;ii:1461–1462. [PMC free article: PMC2079894] [PubMed: 13209151]
  • Kerckaert G.A., Brauninger R., LeBoeuf R.A., Isfort R.J. Use of the Syrian hamster embryo cell transformation assay for carcinogenicity prediction of chemicals currently being tested by the National Toxicology Program in rodent bioassays. Environ. Health Perspectives. 1996;104:1075–1084. [PMC free article: PMC1469685] [PubMed: 8933057]
  • Kune G.A. Laxative use not a risk for colorectal cancer: Data from the Melbourne Colorectal Cancer Study. Z. Gastroenterol. 1993;31:140–143. [PubMed: 8465555]
  • Lambrianides A.L., Rosin R.D. Acute pancreatitis complicating excessive intake of phenolphthalein. Postgrad. med. J. 1984;60:491–492. [PMC free article: PMC2417943] [PubMed: 6463003]
  • LaRusso N.F., McGill D.B. Surreptitious laxative ingestion. Delayed recognition of a serious condition: A case report. Mayo Clin. Proc. 1975;50:706–708. [PubMed: 1195781]
  • Longnecker M.P., Sandler D.P., Haile R.W., Sandler R.S. Phenolphthalein-containing laxative use in relation to adenomatous colorectal polyps in three studies. Environ. Health Perspectives. 1997;105:1210–1212. [PMC free article: PMC1470333] [PubMed: 9370521]
  • Medical Economics (1998) PDR®: Physicians’ Desk Reference, 52nd Ed., Montvale, NJ, Medical Economics Data Production, pp. 2601–2602, 2979.
  • Mehendale H.M. Assessment of hepatobiliary function with phenolphthalein and phenolphthalein glucuronide. Clin. Chem. Enzyme Commun. 1990;2:195–204.
  • Millburn P., Smith R.L., Williams R.T. Biliary excretion of foreign compounds. Biphenyl, stilboestrol and phenolphthalein in the rat: Molecular weight, polarity and metabolism as factors in biliary excretion. Biochem. J. 1967;105:1275–1281. [PMC free article: PMC1198451] [PubMed: 16742556]
  • Mortelmans K., Haworth S., Lawlor T., Speck W., Tainer B., Zeiger E. Salmonella mutagenicity tests. II. Results from the testing of 270 chemicals. Environ. Mutag. 1986;8 Suppl. 7:1–119. [PubMed: 3516675]
  • Mrvos R., Swanson-Biearman B., Dean B.S., Krenzelok E.P. Acute phenolphthalein ingestion in children. A retrospective review. J. pediatr. Health Care. 1991;5:147–151. [PubMed: 1674527]
  • National Toxicology Program (1996) Toxicology and Carcinogenesis Studies of Phenolphthalein (CAS No. 77-09-8) in F344/N Rats and B6C3F1 Mice (Feed Studies) (Technical Report Series No. 465; NIH Publ. No. 97-3390), Research Triangle Park, NC. [PubMed: 12579199]
  • National Toxicology Program (1999) NTP Report on Carcinogens. Background Document for Phenolphthalein (Final, March 1999), Research Triangle Park, NC, NTP Board of Scientific Counselors.
  • Nieto A., Garciá C., López de Haro S. In vivo estrogenic and antiestrogenic activity of phenolphthalein and derivative compounds. Biochem. int. 1990;2:305–311. [PubMed: 2403370]
  • Nishikawa J. Effects of sodium picosulfate and other laxatives in cultured Chang cells. Arzneimittelforschung. 1981;31:1872–1875. [PubMed: 7198466]
  • Parker R.J., Hirom P.C., Millburn P. Enterohepatic recycling of phenolphthalein, morphine, lysergic acid diethylamide (LSD) and diphenylacetic acid in the rat. Hydrolysis of glucuronic acid conjugates in the gut lumen. Xenobiotica. 1980;10:689–703. [PubMed: 7445530]
  • Pietrusko R.G. Use and abuse of laxatives. Am. J. Hosp. Pharm. 1977;34:291–300. [PubMed: 324272]
  • Pohl A., Lowe J.P. Phenolphthalein poisoning—four cases. Proc. Mine med. Off. Assoc. S.A. 1978;57:84–86. [PubMed: 551420]
  • Pouchert, C.J. (1981) The Aldrich Library of Infrared Spectra, 3rd Ed., Milwaukee, WI, Aldrich Chemical Co., p. 1471.
  • Pouchert, C.J. (1985) The Aldrich Library of FT-IR Spectra, Vol. 1-2-3, Milwaukee, WI, Aldrich Chemical Co., p. 1006.
  • Ravdin P.M., van Beurden M., Jordan V.C. Estrogenic effects of phenolphthalein on human breast cancer cells in vitro. Breast Cancer Res. Treat. 1987;9:151–154. [PubMed: 3620717]
  • Rote Liste Sekretariat (1998) Rote Liste 1998, Frankfurt, Rote Liste Service GmbH, pp. 56–068.
  • Royal Pharmaceutical Society of Great Britain (1999) Martindale, The Extra Pharmacopoeia, 13th Ed., London, The Pharmaceutical Press [MicroMedex Online: Health Care Series]
  • Sadtler Research Laboratories (1980) Sadtler Standard Spectra, 1980 Cumulative Index, Philadelphia, PA, p. 1048.
  • Sarcinelli L., Signore L., Malizia E. Lethal phenolphthalein poisoning in a child. Proc. Eur. Soc. Study Drug Toxicol. 1970;50:261.
  • Shelley W.B., Schlappner O.L.A., Heiss H.B. Demonstration of intercellular immunofluorescence and epidermal hysteresis in bullous fixed drug eruption due to phenolphthalein. Br. J. Dermatol. 1972;86:188–125. [PubMed: 4259600]
  • Sidhu P.S., Wilkinson M.L., Sladen G.E., Filipe M.I., Toseland P.A. Fatal phenolphthalein poisoning with fulminant hepatic failure and disseminated intravascular coagulation. Hum. Toxicol. 1989;8:381–384. [PubMed: 2807307]
  • Sipe H.J. Jr, Corbett J.T., Mason R.P. In vitro free radical metabolism of phenolphthalein by peroxidases. Drug Metab. Dispos. 1997;25:468–480. [PubMed: 9107547]
  • Steigmann F., Barnard R.D., Dyniewicz J.M. Phenolphthalein studies: Phenolphthalein in jaundice. Am. J. med. Sci. 1938;196:673–688.
  • Stroud M.B., Rosio T.J. A case of recurring painful red macules. Arch. Dermatol. 1987;123:1225–1230. [PubMed: 2957963]
  • Sugar J.A., Belfer M., Israel E., Herzog D.B. A 3-year-old boy’s chronic diarrhea and unexplained death. J. Am. Acad. Child adolesc. Psychiatr. 1991;30:1015–1021. [PubMed: 1757428]
  • Sund R.B., Hillestad B. Uptake, conjugation and transport of laxative diphenols by everted sacs of the rat jejunum and stripped colon. Acta pharmacol. toxicol. 1982;51:377–387. [PubMed: 6897480]
  • Sund R.B., Lauterbach F. Drug metabolism and metabolite transport in the small and large intestine: Experiments with 1-naphthol and phenolphthalein by luminal and contraluminal administration in the isolated guinea pig mucosa. Acta pharmacol. toxicol. 1986;58:74–83. [PubMed: 3953296]
  • Sund R.B., Hetland H.S., Nafstad I. Autoradiography in mice after intravenous and intragastric administration of phenolphthalein and desacetylated bisacodyl, two laxative diphenols of the diphenylmethane group. Norw. pharm. Acta. 1986;48:57–73.
  • Swiss Pharmaceutical Society, ed. (1999) Index Nominum, International Drug Directory, 16th Ed., Stuttgart, Medpharm Scientific Publishers [MicroMedex Online: Health Care Series]
  • Thomas, J., ed. (1998) Australian Prescription Products Guide, 27th Ed., Victoria, Australian Pharmaceutical Publishing, Vol. 1, pp. 286, 1190–1191, 1229, 1587, 1686.
  • Tice R.R., Furedi-Machacek M., Satterfield D., Udumudi A., Vasquez M., Dunnick J.K. Measurement of micronucleated erythrocytes and DNA damage during chronic ingestion of phenolphthalein in transgenic female mice heterozygous for the p53 gene. Environ. mol. Mutag. 1998;31:113–124. [PubMed: 9544189]
  • Tsutsui T., Tamura Y., Yagi E., Hasegawa K., Tanaka Y., Uehama A., Someya T., Hamaguchi F., Yamamoto H., Barrett J.C. Cell-transforming activity and genotoxicity of phenolphthalein in cultured Syrian hamster embryo cells. Int. J. Cancer. 1997;73:697–701. [PubMed: 9398048]
  • Tyson R.M., Shrader E.A., Perlman H.H. Drugs transmitted through breast milk. Part I. Laxatives. J. Pediatr. 1937;11:824.
  • US Pharmacopeial Convention (1994) The 1995 US Pharmacopeia, 23rd Rev./The National Formulary, 18th Rev., Rockville, MD, pp. 1205–1206.
  • US Pharmacopeial Convention (1998) USP Dispensing Information, Drug Information for the Health Care Professional, 18th Ed., Rockville, MD, Vol. I, pp. 1798–1840.
  • Velentzas C.G., Ikkos D.G. Phenolphthalein as cause of factitious enteritis (Letter to the Editor). J. Am. med. Assoc. 1971;217:966. [PubMed: 5109372]
  • Visek W.J., Liu W.C., Roth L.J. Studies on the fate of carbon-14 labeled phenolphthalein. J. Pharmacol. exp. Ther. 1956;117:347–357. [PubMed: 13332581]
  • Weiss B.D., Wood G.A. Laxative abuse causing gastrointestinal bleeding. J. Fam. Pract. 1982;15:177–181. [PubMed: 6979610]
  • WHO (1997) Alert No. 65: Laxatives Containing Phenolphthalein—Proposed Ban on Sale of OTC Products: Risk of Carcinogenicity [http://www​.who.int/dmp​/drugalert/alert65.htm]
  • WHO, author. Regulatory matters: Phenolphthalein products withdrawn. WHO Drug Inf. 1998;12:13–14.
  • Williams, R.T. (1959) Detoxication Mechanisms. The Metabolism and Detoxication of Drugs, Toxic Substances and Other Organic Compounds, 2nd Ed., New York, John Wiley & Sons, pp. 475–476.
  • Witt K.L., Gulati D.K., Kaur P., Shelby M.D. Phenolphthalein: Induction of micro-nucleated erythrocytes in mice. Mutat. Res. 1995;341:151–160. [PubMed: 7529356]
  • Wyatt E., Greaves M., Søndergaard J. Fixed drug eruption (phenolphthalein). Evidence for a blood-borne mediator. Arch. Dermatol. 1972;106:671–673. [PubMed: 4117910]
  • Zanolli M.D., McAlvany J., Krowchuk D.P. Phenolphthalein-induced fixed drug eruption: A cutaneous complication of laxative use in a child. Pediatrics. 1993;91:1199–1201. [PubMed: 8502530]

Vitamin K substances

Vitamin K comprises a group of substances, which are widespread in nature and are an essential co-factor in humans in the synthesis of several proteins that play a role in haemostasis and others that may be important in calcium homeostasis. The K vitamins all contain the 2-methyl-1,4-naphthoquinone (menadione) moiety, and the various naturally occurring forms differ in the alkyl substituent at the 3-position. Phylloquinone (vitamin K1) is 2-methyl-3-phytyl-1,4-naphthoquinone and is widely found in higher plants, including green leafy vegetables, and in green and blue algae. The menaquinones (formerly vitamin K2) have polyisoprenyl substituents at the 3-position and are produced by bacteria. The compound menadione (formerly vitamin K3) lacks an alkyl group at the 3-position but can be alkylated in vivo in some species. Several synthetic water-soluble derivatives, such as the sodium diphosphate ester of menadiol and the addition product of menadione with sodium bisulfite, also have commercial applications (National Research Council, 1989; Gennaro, 1995; Weber & Rüttimann, 1996).

1. Exposure Data

1.1. Chemical and physical data

1.1.1. Nomenclature, structural and molecular formulae and relative molecular
Vitamin K (generic)
  • Chem. Abstr. Serv. Reg. No.: 12001-79-5
  • Chem. Abstr. Name: Vitamin K
Vitamin K1 (generic)
  • Chem. Abstr. Serv. Reg. No.: 11104-38-4
  • Chem. Abstr. Name: Vitamin K1
Phylloquinone
  • Chem. Abstr. Serv. Reg. No.: 84-80-0
  • Deleted CAS Reg. Nos.: 10485-69-5; 15973-57-6; 50926-17-5
  • Chem. Abstr. Name: 2-Methyl-3-[(2E,7R,11R)-3,7,11,15-tetramethyl-2-hexadecenyl]-1,4-naphthalenedione
  • IUPAC Systematic Name: [R-[R*,R*-(E)]]-2-Methyl-3-(3,7,11,15-tetramethyl-2-hexadecenyl)-1,4-naphthalenedione
  • Synonyms: Antihaemorrhagic vitamin; 2-methyl-3-phytyl-1,4-naphthoquinone; 2-methyl-3-(3,7,11,15-tetramethyl-2-hexadecenyl)-1,4-naphthalenedione; α-phylloquinone; trans-phylloquinone; phylloquinone K1; phytomenadione; phytonadione; phytylmenadione; 3-phytylmenadione; phytylmenaquinone; vitamin K1; vitamin K1(20); 2′,3′-trans-vitamin K1 [Note: The IUPAC recommends use of the name ‘phylloquinone’ and the abbreviation ‘K’ (rather than ‘K1’). Both phylloquinone and vitamin K1 are in common use. The United States Pharmacopeia uses the name ‘phytonadione’; The European Pharmacopoeia uses the name ‘phytomenadione’, which is a synonym occasionally found in the pharmaceutical and pharmacological literature.]
Image 418a
Menaquinone-4
  • Chem. Abstr. Serv. Reg. No.: 863-61-6
  • Deleted CAS Reg. Nos.: 15261-37-7; 20977-31-5; 39776-41-5
  • Chem. Abstr. Name: 2-Methyl-3-[(2E,6E,10E)-3,7,11,15-tetramethyl-2,6,10,14-hexadecatetraenyl]-1,4-naphthalenedione
  • IUPAC Systematic Name: 2-Methyl-3-(3,7,11,15-tetramethyl-2,6,10,14-hexadecatetraenyl)-1,4-naphthoquinone
  • Synonyms: Menaquinone-K4; menatetrenone; (E,E,E)-2-methyl-3-(3,7,11,15-tetramethyl-2,6,10,14-hexadecatetraenyl]-1,4-naphthalenedione; MK4; vitamin K2(20); vitamin MK4
Image 418b
Vitamin K2 (generic)
  • Chem. Abstr. Serv. Reg. No.: 11032-49-8
  • Chem. Abstr. Name: Vitamin K2
Menadione
  • Chem. Abstr. Serv. Reg. No.: 58-27-5
  • Chem. Abstr. Name: 1,4-Naphthalenedione, 2-methyl-
  • IUPAC Systematic Name: 1,4-Naphthoquinone, 2-methyl-
  • Synonyms: 1,4-Dihydro-1,4-dioxo-2-methylnaphthalene; 2-methyl-1,4-naphthalenedione; 2-methylnaphthoquinone; β-methyl-1,4-naphthoquinone; 2-methyl-1,4-naphthoquinone; 3-methyl-1,4-naphthoquinone; MK-0; vitamin K0; vitamin K2(0); vitamin K3 [Note: ‘Menadione’ is the common name preferred by IUPAC for the chemical, previously called vitamin K3]
Image 419a
Menadione sodium bisulfite
  • Chem. Abstr. Serv. Reg. No.: 130-37-0
  • Alternate CAS Reg. No.: 57414-02-5
  • Deleted CAS Reg. Nos.: 8012-53-1; 8017-97-8; 8028-24-8; 8053-08-5
  • Chem. Abstr. Name: 1,2,3,4-Tetrahydro-2-methyl-1,4-dioxo-2-naphthalenesulfonic acid, sodium salt
  • IUPAC Systematic Name: 1,2,3,4-Tetrahydro-2-methyl-1,4-dioxo-2-naphthalenesulfonic acid, sodium salt
  • Synonyms: 3,3-Dihydro-2-methyl-1,4-naphthoquinone-2-sulfonate sodium; menadione sodium hydrogen sulfite; menaphthone sodium bisulfite; menaphthone sodium bisulphite; 2-methyl-1,4-naphthalenedione, sodium bisulfite deriv.; 2-methyl-1,4-naphthoquinone sodium bisulfite; 2-methylnaphthoquinone sodium hydrogen sulfite; 2-methyl-1,4-naphthoquinone sodium hydrogen sulfite; MSBC; sodium menadione bisulfite; vitamin K injection; vitamin K3 sodium bisulfite
Image 420a
Menadione sodium bisulfite trihydrate
  • Chem. Abstr. Serv. Reg. No.: 6147-37-1
  • Chem. Abstr. Name: 1,2,3,4-Tetrahydro-2-methyl-1,4-dioxo-2-naphthalenesulfonic acid, sodium salt, trihydrate
Image 420b
Menadiol
  • Chem. Abstr. Serv. Reg. No.: 481-85-6
  • Chem. Abstr. Name: 2-Methyl-1,4-naphthalenediol
  • IUPAC Systematic Name: 2-Methyl-1,4-naphthalenediol
  • Synonyms: Dihydrovitamin K3; menaquinol; 2-methyl-1,4-dihydroxynaphthalene; 2-methylhydronaphthoquinone; 2-methylnaphthalene-1,4-diol; 2-methyl-1,4-naphthohydroquinone; 2-methyl-1,4-naphthoquinol; reduced menadione; reduced vitamin K3; vitamin K3H2
Image 420c
Menadiol sodium phosphate
  • Chem. Abstr. Serv. Reg. No.: 131-13-5
  • Chem. Abstr. Name: 2-Methyl-1,4-naphthalenediol, bis(dihydrogen phosphate), tetrasodium salt
  • IUPAC Systematic Name: 2-Methyl-1,4-naphthalenediol, diphosphate, tetrasodium salt
  • Synonyms: Menadiol diphosphate tetrasodium salt; menadiol sodium diphosphate; menadiol tetrasodium diphosphate; menadione diphosphate tetrasodium salt; 2-methyl-1,4-naphthoquinol bis(disodium phosphate); tetrasodium 2-methyl-1,4-naphthalenediol bis(dihydrogen phosphate)
Image 421a
Menadiol sodium phosphate hexahydrate
  • Chem. Abstr. Serv. Reg. No.: 6700-42-1
  • Chem. Abstr. Name: 2-Methyl-1,4-naphthalenediol, bis(dihydrogen phosphate), tetrasodium salt, hexahydrate
  • IUPAC Systematic Name: 2-Methyl-1,4-naphthalenediol, diphosphate, tetrasodium salt, hexahydrate
  • Synonyms: Menadiol sodium diphosphate hexahydrate
Image 421b
Acetomenaphthone
  • Chem. Abstr. Serv. Reg. No.: 573-20-6
  • Chem. Abstr. Name: 2-Methyl-1,4-naphthalenediol, diacetate
  • IUPAC Systematic Name: 2-Methyl-1,4-naphthalenediol, diacetate
  • Synonyms: 1,4-Diacetoxy-2-methylnaphthalene; menadiol diacetate; 2-methyl-1,4-naphthohydroquinone diacetate; 2-methyl-1,4-naphthoquinol diacetate; 2-methyl-1,4-naphthylene diacetate; vitamin K diacetate; vitamin K4
Image 422a

IUPAC recommends that 2-methyl-3-polyprenyl-1,4-naphthoquinone be referred to as menaquinone-n, previously vitamin K2, n being the number of prenyl residues. Vitamin K2(20) is so named because it contains 20 carbon atoms in the chain. In the biological literature, vitamin K2 is frequently referred to as menaquinone and is further designated by the number of isoprene units in the side-chain. For example, vitamin K2(20) is also called menaquinone-4 for the four isoprene units in the side-chain. The compound originally isolated from rotting fish meal and named vitamin K2 was later identified as menaquinone-7 (2-methyl-3-farnesylgeranyl-geranyl-1,4-naphthoquinone). In the older literature, the designation vitamin K2(35) is used for menaquinone-7, but this is no longer used. Menaquinones found in nature have side-chains of 4–13 isoprenoid residues and are usually in the all-trans configuration; however, menaquinones with the cis configuration and partially saturated side-chains also exist (Suttie, 1985, 1991; Weber & Rüttimann, 1996; Van Arnum, 1998).

1.1.2. Chemical and physical properties of the pure substances
Phylloquinone
  • (a) Description: Clear, yellow to amber, very viscous, odourless liquid (Gennaro, 1995; Budavari, 1996)
  • (b) Spectroscopy data: Ultraviolet, infrared, nuclear magnetic resonance (proton and 13C) and mass spectral data have been reported (Hassan et al., 1988).
  • (c) Solubility: Insoluble in water; sparingly soluble in methanol; soluble in acetone, benzene, chloroform, diethyl ether, dioxane, ethanol, hexane, petroleum ether and other fat solvents and vegetable oils (Budavari, 1996)
  • (d) Stability: Stable to air and moisture; decomposes in sunlight; unaffected by dilute acids; destroyed by solutions of alkali hydroxides and by reducing agents (Gennaro, 1995; Budavari, 1996)
  • (e) Optical rotation: [α]25d, −28° (Budavari, 1996)
Menaquinone-4

From Japan Medical Products Trade Association (1996)

  • (a) Description: Yellow crystals or an oily substance
  • (b) Melting-point: 34–38 °C
  • (c) Solubility: Practically insoluble in water; very soluble in diethyl ether, chloroform and hexane; freely soluble in isooctane; sparingly soluble in ethanol and isopropanol; slightly soluble in methanol
  • (d) Stability: Decomposed by light or alkalis
Menadione
  • (a) Description: Bright-yellow crystals with a very faint acrid odour (Budavari, 1996)
  • (b) Melting-point: 105–107 °C (Budavari, 1996)
  • (c) Spectroscopy data: Infrared (prism [8077]; grating [8522]), ultraviolet [2183] and nuclear magnetic resonance (proton [3217]; 13C [6002]) spectral data have been reported (Sadtler Research Laboratories, 1980; British Pharmacopoeial Commission, 1993).
  • (d) Solubility: Insoluble in water; soluble in benzene (1 g/10 mL), ethanol (1 g/60 mL), and vegetable oils (1 g/50 mL); moderately soluble in carbon tetrachloride and chloroform (Budavari, 1996)
  • (e) Stability: Stable in air; decomposed by sunlight; destroyed by alkalis and reducing agents (Budavari, 1996)
Menadione sodium bisulfite (trihydrate)
  • (a) Description: White, crystalline, odourless, hygroscopic powder (Gennaro, 1985; Budavari, 1996)
  • (b) Solubility: Soluble in water (∼0.5 g/mL); slightly soluble in chloroform and ethanol; practically insoluble in benzene and diethyl ether (Gennaro, 1985; Budavari, 1996)
  • (c) Stability: Discolours and may turn purple under light (Budavari, 1996)
Menadiol
Menadiol sodium phosphate (hexahydrate)
Acetomenaphthone
  • (a) Description: Crystalline solid (Budavari, 1996)
  • (b) Melting-point: 112–114 °C (Budavari, 1996)
  • (c) Solubility: Practically insoluble in water; slightly soluble in ethanol; soluble in acetic acid (Budavari, 1996)
  • (d) Spectroscopy data: Infrared (prism [20206]; grating [32489]), ultraviolet [6761] and nuclear magnetic resonance (proton [2298]; 13C [2451]) spectral data have been reported (Sadtler Research Laboratories, 1980).
1.1.3. Technical products and impurities

Commercially available phylloquinone is prepared synthetically and may contain not only 2′,3′-trans-phylloquinone (not less than 75%) but also 2′,3′-cis-phylloquinone and trans-epoxyphylloquinone (not more than 4.0%). Phylloquinone occurs in nature only as the 2′,3′-trans-phylloquinone stereoisomer (Weber & Rüttimann, 1996; American Hospital Formulary Service, 1997; Council of Europe, 1997).

Phylloquinone is available as a 5- and 10-mg tablet (chewable), a 2- and 10 mg/mL injection solution, a 10- and 20-mg/mL oral solution and a 20-mg/mL emulsion. The tablet may also contain carmellose, carob bean flour, carob gum, cocoa butter, cocoa powder, ethyl cellulose, ethyl vanillin, glucose, glycerol, gum arabic, hard and viscous paraffin, lactose, rice starch, sugar, silicic acid, silicon dioxide, skim-milk powder, sodium cyclamate, talc and titanium dioxide. The injection solution may also contain benzyl alcohol, dextrose, glacial acetic acid, glucose, glycocholic acid, hydrochloric acid, macrogol ricinoleate, phenol, phosphatidylcholine from soya beans, polyethoxylated fatty acid derivative (castor oil), polysorbate 80, propylene glycol, sodium acetate, sodium hydroxide and water. A widely used injectable formulation, Konakion®, formerly contained a polyethoxylated castor oil as an emulsifying agent, but has been reformulated as a mixed micellar preparation, Konakion MM®, containing glycocholic acid, lecithin and buffered to pH 6. The oral solution may also contain benzoic acid, glycocholic acid, hydrochloric acid, lecithin, macrogol ricinoleate, methyl 4-hydroxybenzoate, propyl 4-hydroxybenzoate, sodium hydroxide and water. The emulsion may also contain polysorbate 80, purified water and sorbic acid. Phylloquinone is also available as a component (200 µg) of a multivitamin lyophilized, sterile powder intended for reconstitution and dilution in intravenous infusions, as a component (0.075 mg) of an effervescent multivitamin tablet, and as a component (5.5 µg) of a multivitamin infant formula (Gennaro, 1995; American Hospital Formulary Service, 1997; Canadian Pharmaceutical Association, 1997; British Medical Association/Royal Pharmaceutical Society of Great Britain, 1998; Editions du Vidal, 1998; LINFO Läkemedelsinformation AB, 1998; Rote Liste Sekretariat, 1998; Thomas, 1998; US Pharmacopeial Convention, 1998).

Trade names for phylloquinone include AquaMEPHYTON, AquaMephyton, AquaMephyton R, Combinal K1, Hymeron, Kanakion, Kanavit, Kaywan, Kephton, Kinadion, K1 Delagrange, Konakion, Konakion MM, Menadion ‘Dak’, Mephyton, Monodion, Synthex P, Vitacon, Vita-K1, Vitamina K1 Biol, Vitamine K1 Roche and Vitamin K1 (CIS Information Services, 1998; Royal Pharmaceutical Society of Great Britain, 1999; Swiss Pharmaceutical Society, 1999).

Menaquinone-4 is available in Japan as 5- and 15-mg capsules and as a 2-mg/mL syrup. The capsules may also contain ethyl parahydroxybenzoate, propyl paraoxyhydroxybenzoate, sodium lauryl sulfate and FD&C Yellow No. 6 (Sunset Yellow). The syrup may also contain polyoxyethylene hydrogenated castor oil 60, propylene glycol, ethyl parahydroxybenzoate, sodium benzoate and flavouring (Japan Medical Products Trade Association, 1996).

Trade names for menaquinone-4 include Glakay and Kaytwo (Japan Medical Products Trade Association, 1996).

Menadione is available as a 2-, 5- and 10-mg tablet and as a 2- and 10-mg/mL injection (in oil). Menadione sodium bisulfite is available as a 10-mg tablet and as a 5- and 10-mg/mL and 72-mg/10 mL injection (Gennaro, 1985).

Trade names for menadione include Aquakay, Aquinone, Austrovit-K Depot, Hemodal, K-Thrombyl, K-Vitan, Kaergona, Kanone, Kaom Belgarum, Kappaxan, Kappaxin, Karanum, Karcon, Kareon, Kativ-G, Kavitamin, Kayklot, Kaykot, Kayquinone, Kipca, Kipca-Oil Soluble, Klottone, Koaxin, Kolklot, Menadion, Menaphthon, Menaphthone, Menaquinone 0, Mitenon, Mitenone, MNQ, Neo-Zimema-K, Panosine, Prokayvit, Synkay, Thyloquinone, Vikaman, Vita-Noxi K and Vitavel-K (Swiss Pharmaceutical Society, 1999).

Trade names for menadione sodium bisulfite include Austrovit-K, Golagen K, Hemoklot, Hetrogen K, Hetrogen K Premix, Hykinone, Ido-K, K-Thrombin, K-Trombina, Kalzon, Kareon, Kavitamin, Kavitan, Kavitol, Kawitan, Klotogen, Libavit K, Nuvit K, Vikaman, Vikasol, Vitaminum K and Zimema K (Swiss Pharmaceutical Society, 1999).

Menadiol sodium phosphate (as the hexahydrate) is available as a 5-mg and 10 mg (equivalent of menadiol phosphate) tablet and as 5- and 10-mg/mL and 75-mg/2 mL injections (Gennaro, 1995; British Medical Association/Royal Pharmaceutical Society of Great Britain, 1998; US Pharmacopeial Convention, 1998).

Acetomenaphthone is available in a chilblain formula tablet containing 30 mg nicotinamide and 5 mg acetomenaphthone and as a component (10 mg) of a multivitamin injection solution, which may also contain butyl hydroxyanisole, butyl hydroxytoluene, peanut oil, medium-chain triglycerides and olive oil (Rote Liste Sekretariat, 1998; Thomas, 1998).

Trade names for menadiol sodium phosphate hexahydrate include Kappadione, Kativ (injection), Kipca water soluble, Naphthidone, Procoagulo, Synkavit, Synka-Vit, Synkavite, Synkayvite and Thylokay (Swiss Pharmaceutical Society, 1999).

Trade names for acetomenaphthone include Adaprin, Davitamon-K, Davitamon-Koral, Kapathrom, Kapilin, Kapilon, Kappaxan, Kativ powder, Kayvite, Pafavit, Prokayvit Oral and Vitavel K.

1.1.4. Analysis

Several international pharmacopoeias specify infrared (IR) and ultraviolet (UV) absorption spectrophotometry with comparison to standards as the methods for identifying phylloquinone; UV absorption spectrophotometry and liquid chromatography are used to assay its purity. Phylloquinone is identified in pharmaceutical preparations by IR and UV absorption spectrophotometry and liquid chromatography; liquid chromatography is used to assay for its content (British Pharmacopoeial Commission, 1993; US Pharmacopeial Convention, 1994; Society of Japanese Pharmacopoeia, 1996; Council of Europe, 1997). AOAC International (1996) has developed a liquid chromatographic method with UV detection for the determination of phylloquinone in ready-to-feed milk-based infant formulae.

As a result of its high selectivity and sensitivity, high-performance liquid chromatography (HPLC) is the method of choice for the determination of phylloquinone and menaquinones in blood, tissues, milk and foods. Various procedures for extraction and preliminary purification, normal or reversed-phase HPLC and UV, electrochemical and fluorescence detection (both after electrochemical or chemical reduction and after photochemical decomposition) of the various vitamin K substances have been described. The limit of detection of phylloquinone is 25–500 pg, depending on the detection method used. Similar values, which vary according to the length of the side-chain, apply to the menaquinones. HPLC methods are also available for the determination of menadione and water-soluble derivatives in feedstuffs, premixes and vitamin concentrates (Weber & Rüttimann, 1996).

Alternative methods are thin-layer chromatography, high-performance thin-layer chromatography and gas chromatography. The spectrophotometric, fluorimetric and colorimetric methods previously used without chromatographic purification of the samples to be analysed are frequently less sensitive and less specific than HPLC, for instance allowing no distinction between phylloquinone and menaquinones (Weber & Rüttimann, 1996).

Several international pharmacopoeias specify IR absorption spectrophotometry with comparison to standards and colorimetry as the methods for identifying menadiol sodium phosphate hexahydrate; potentiometric titration with ceric sulfate is used to assay its purity. In pharmaceutical preparations, menadiol sodium phosphate is identified by IR absorption spectrophotometry and colorimetry; potentiometric titration with ceric sulfate and UV absorption spectrophotometry are used to assay for its content (British Pharmacopoeial Commission, 1993; US Pharmacopeial Convention, 1994; Council of Europe, 1997).

Several international pharmacopoeias specify IR and UV absorption spectrophotometry with comparison to standards as the methods for identifying menadione; titration with ammonium and cerium nitrate or ceric sulfate is used to assay its purity. Visible (635 nm) absorption spectrophotometry is used to assay for its content in pharmaceutical preparations (British Pharmacopoeial Commission, 1993; US Pharmacopeial Convention, 1994; Council of Europe, 1997).

1.2. Production

Although the predominant commercial form of phylloquinone is the synthetic race-mate, natural phylloquinone is accessible either by extraction from a natural source or from condensation of menadione with natural phytol. The stability of phylloquinone to heat made possible the use of commercially dehydrated alfalfa meal, for example, as a natural source (Hassan et al., 1988). The synthesis and spectral properties of all four stereoisomers of (E)-phylloquinone have been described and their biological potencies determined. When natural phylloquinone was used as a standard in bioassays, it was concluded that all four stereoisomers have essentially identical activity (Van Arnum, 1998).

The first syntheses and structural elucidation of phylloquinone were published in 1939 almost simultaneously by four groups. The starting materials were menadione or menadiol as the aromatic component and natural phytol or one of its derivatives. A breakthrough in commercial synthesis was achieved in the 1950s, when it was found that monoacylated menadiols (e.g. the monoacetate or the monobenzoate) could be used advantageously in the alkylation step and that natural phytol could be replaced by isophytol, which is easy to synthesize (Weber & Rüttimann, 1996).

In the Isler-Lindlar method, excess menadiol monobenzoate is condensed with isophytol in the presence of boron trifluoride etherate as a catalyst. The alkylation product is obtained as a 70:30 trans/cis mixture. The trans form can be enriched by recrystallization. The trans-enriched alkylation product (trans:cis 9:1) is saponified with potassium hydroxide and oxidized to phylloquinone with oxygen (Weber & Rüttimann, 1996).

The industrial synthesis of menaquinones parallels that of phylloquinone and involves as a key step alkylation of monosubstituted menadione with an appropriate (all-trans) polyisoprenyl derivative. Considerably more work has been done on fermentative approaches to menaquinones than for phylloquinone. Menaquinones of varying chain lengths, from C5 to C65, have been produced and isolated from bacteria. Menaquinone-4 is produced and used extensively in Japan (Van Arnum, 1998).

Menadione can be prepared by oxidizing 2-methylnaphthalene with chromic acid or hydrogen peroxide (Weber & Rüttimann, 1996). A process based on biotechnological techniques has been reported in Japan (Van Arnum, 1998).

Menadione sodium bisulfite can be prepared by reacting menadione with sodium bisulfite. The reaction may be visualized as consisting of the typical addition of sodium bisulfite to a ketone, forming the R(OH)(SO3Na) compound, which then rearranges at the expense of one degree of unsaturation of the quinoid nucleus. The compound readily regenerates menadione on treatment with mild alkali and behaves as a typical ketone–sodium bisulfite addition compound (Gennaro, 1985; Van Arnum, 1998).

Menadiol sodium phosphate can be prepared by reducing menadione to the diol, followed by double esterification with hydriodic acid, metathesis of the resulting 1,4-diiodo compound with silver phosphate and neutralization of the bis(dihydrogen phosphate) ester with sodium hydroxide (Gennaro, 1995).

Information available in 1999 indicated that phylloquinone was manufactured and/or formulated in 41 countries, menadione in 26 countries, menadione sodium bisulfite in 21 countries, menadiol and menadiol sodium phosphate (as the hexahydrate) in two countries each and acetomenaphthone in seven countries (CIS Information Services, 1998; Royal Pharmaceutical Society of Great Britain; 1999; Swiss Pharmaceutical Society, 1999).

1.3. Use

1.3.1. Physiological function

The only established biochemical role for vitamin K is as a cofactor in a unique post-translational chemical modification in which selective glutamate (Glu) residues on certain specialized calcium-binding proteins are transformed to γ-carboxyglutamate (Gla) residues (Suttie, 1991; Shearer, 1997). The modification is catalysed by a microsomal enzyme called γ-glutamyl or vitamin K-dependent carboxylase, which is present in most tissues. The best-known vitamin K-dependent proteins are those synthesized in the liver, which play a role in the maintenance of normal haemostasis. They comprise four proteins (II, VII, IX and X) that promote coagulation and two proteins (C and S) that act in the regulatory feedback control of coagulation. Vitamin K-dependent proteins, of uncertain function, are also known to occur in a variety of other tissues such as bone, kidney, pancreas, placenta, spleen and lungs. They include the bone protein osteocalcin (also called bone Gla protein) and matrix Gla protein; there is growing evidence that these proteins may be important for bone health and other regulatory functions in calcium metabolism. In those proteins with well-established functions, such as coagulation proteins, the Gla groups are essential for the biological activity (Thijssen & Drittij-Reijnders, 1996; Shearer, 1997).

Naturally occurring phylloquinone and menaquinones all γ-carboxylate the vitamin K-dependent coagulation proteins. Synthetic forms of menadione (and related water-soluble salts) that lack a side-chain at the 3-position have biological activity in vivo only after side-chain alkylation, which results in the specific synthesis of menaquinone-4 (Suttie, 1991; see also section 4).

1.3.2. Supplementation and therapy

Vitamin K is given as a supplement to prevent or cure vitamin K deficiency when the endogenous vitamin K supply from the diet is likely to become or has proven to be insufficient. Neonates are born with very limited vitamin K stores, but most infants do not show relevant hypoprothrombinaemia at birth (von Kries et al., 1987a, 1988; von Kries, 1991). Biochemical signs of vitamin K deficiency are common during the first week of life, however, unless sufficient amounts of vitamin K are ingested. The natural diet of newborns is human milk, which contains vitamin K at concentrations of 0.69–9.2 ng/mL (see Table 1). [The Working Group noted that some of the high values in the Table may reflect methodological problems with analysis and milk collection.] Bleeding, the classical clinical manifestation of vitamin K deficiency, is extremely rare on the first day of life, and the typical time of onset is during the first week, with bleeding from mucous membranes, the umbilicus, following circumcision, and rarely, into the central nervous system (von Kries et al., 1988; von Kries, 1991). This condition was originally called ‘classical haemorrhagic disease of the newborn’; the present nomenclature is ‘classical vitamin K deficiency bleeding’ (Sutor et al., 1999).

Table 1. Concentrations of phylloquinone in human and cow’s milk, infant formulae and various oils.

Table 1

Concentrations of phylloquinone in human and cow’s milk, infant formulae and various oils.

During the first three months of life, exclusively breast-fed infants remain at risk for vitamin K deficiency bleeding. In many of these infants, the bleeding episode, which is often intracranial haemorrhage, is the first perceived symptom of an underlying cholestatic disease. In 10–30% of the cases, however, no underlying disease can be found (von Kries et al., 1988).

After the first three months of life, vitamin K deficiency is almost completely confined to patients with cholestatic diseases (congenital or acquired obstruction of the bile duct), malabsorption syndromes or cystic fibrosis (Houwen et al., 1987; van den Anker & Sinaasappel, 1993; O’Brien et al., 1994; Kowdley et al., 1997; Nowak et al., 1997; see also section 1.3.4).

1.3.3. For prevention of vitamin K deficiency in newborns and early infancy

The use of vitamin K prophylaxis since the 1950s has varied widely over time, between countries and within countries between institutions. The predominant patterns were to give either selective intramuscular prophylaxis only to infants presumed to be at special risk for bleeding (mainly premature and low-birth-weight infants and those delivered surgically) or general prophylaxis for all infants. In the latter case, vitamin K was given either intramuscularly or orally.

Several preparations of fat-soluble vitamin K have been in use. In the early 1950s, water-soluble menadiol sodium phosphate was widely used, until haemolysis due to high doses of this preparation in neonates was identified (Meyer & Angus, 1956). In most countries, phylloquinone has been used since that time, although in some third-world countries water-soluble menadione sodium bisulfite still seems to be used (Sharma et al., 1995). Because it is technically difficult to dissolve phylloquinone, only a limited number of preparations became available. The Roche preparation (Konakion®) in which Cremophor (polyethoxylated castor oil) is used as an emulsifying vehicle has been widely available in Europe and North America. The manufacturer has recently replaced the Cremophor preparation by a new mixed micellar preparation Konakion–MM® (British Medical Association/Royal Pharmaceutical Society of Great Britain, 1998). In Japan, an oral preparation of menaquinone-4 is used instead of phylloquinone (Hanawa, 1992).

Almost all cases of vitamin K deficiency bleeding can be prevented by intramuscular administration of 1 mg of vitamin K at birth (von Kries & Hanawa, 1993). Clinical observations and laboratory investigations have also clearly shown that a single oral dose of vitamin K protects against classical vitamin K deficiency bleeding (Clark & James, 1995) but is less effective for prevention of this condition later in life (Tönz & Schubiger, 1988; Ekelund, 1991). Without vitamin K prophylaxis, the incidence of late vitamin K deficiency bleeding in Europe was estimated to be 40–100 per million livebirths, whereas in Asia the condition appears to be considerably more common (Hanawa, 1992; Choo et al., 1994).

Since intramuscular vitamin K prophylaxis has proven effective against late deficiency bleeding, 1 mg of vitamin K at birth was recommended in most western countries (von Kries, 1991). After reports of a potential association between vitamin K prophylaxis and the risk for childhood cancer (Golding et al., 1990, 1992), several countries switched to oral prophylaxis regimens with repeated doses of phylloquinone (Hill, 1994; Doran et al., 1995; Hansen & Ebbesen, 1996; Cornelissen et al., 1997). The optimal oral dose regimen remains to be established (von Kries, 1999).

1.3.4. Cholestatic and malabsorption syndromes

Vitamin K deficiency is observed in patients with cholestatic jaundice, cystic fibrosis, primary biliary cirrhosis and other diseases. In most cases, however, vitamin K deficiency is detectable only by measuring the plasma concentrations of vitamin K or with sensitive biochemical markers of vitamin K deficiency (Cornelissen et al., 1992; O’Brien et al., 1994; Kowdley et al., 1997). Bleeding is observed only rarely. Additional risk factors, such as therapy with antibiotics that interfere with vitamin K metabolism, may cause bleeding in patients with cystic fibrosis (Nowak et al., 1997). Some patients with this disease are given vitamin K supplements, although there are no uniform recommendations (Durie, 1994).

1.3.5. Vitamin supplementation to overcome side-effects of drugs that interfere with vitamin K metabolism

An important indication for vitamin K supplementation is the side-effects of drugs that interfere with its metabolism. Mothers on antiepileptic drugs, for example, are at high risk of delivering an infant with manifest vitamin K deficiency (Cornelissen et al., 1993a) and intracranial bleeding (Renzulli et al., 1998).

Hypoprothrombinaemia may be caused by some cephalosporins, especially those containing an N-methylthiotetrazole side-chain, and may require vitamin K supplementation (Breen & St Peter, 1997).

1.3.6. Vitamin K therapy
(a) Overdosage of vitamin K antagonists

The coumarin derivatives acenocoumarol, phenprocoumon and warfarin are among the most commonly used oral anticoagulants (Keller et al., 1999). The clinical symptom of overdosage of these drugs is bleeding. A tendency to bleed is also increased by individual susceptibility to one of these anticoagulants, interference with other drugs or poor dietary intake of vitamin K. The biochemical indicator for over-dosage is an excessive prolongation of the prothrombin time. Minor bleeding is most commonly managed by temporarily discontinuing treatment and by giving vitamin K to counteract the effects of the coumarin derivative. In the case of major bleeding, especially intracranial haemorrhages, higher doses of vitamin K and use of prothrombin complex concentrates are recommended to induce immediate reversal of anticoagulation (Pindur et al., 1999). In the past, the oral or intravenous dose of phylloquinone used to counteract supratherapeutic anticoagulation was 10–50 mg (Fetrow et al., 1997). Much lower doses have been proposed recently. In asymptomatic patients, a 1-mg oral dose of vitamin K was shown to reduce the international normalized ratio effectively (Crowther et al., 1998). Low subcutaneous doses of phylloquinone are an effective alternative to intravenous administration of phylloquinone in the treatment of warfarin-induced hypoprothrombinaemia (Fetrow et al., 1997).

(b) Prevention of intracranial haemorrhage in very-low-birth-weight, premature infants

The effect of high doses of vitamin K given to women at imminent risk of early preterm parturition has been studied with the primary aim of preventing periventricular haemorrhage and the associated neurological injury in the infant. A first meta-analysis of the trials came to the conclusion, however, that it is ineffective (Thorp et al., 1995).

1.3.7. Other uses

Menadione is of industrial importance as an intermediate in the synthesis of phylloquinone, and salts of its bisulfite adduct are used as stabilized forms in the animal feed industry. Commercially significant forms are menadione sodium bisulfite and menadione dimethyl pyrimidinol (Van Arnum, 1998).

Menaquinone-4 has been used in Japan at high doses for the treatment of osteoporosis (Shearer, 1997).

1.4. Occurrence

Phylloquinone is widely distributed in higher plants and in some blue–green algae. It is present in many foods, especially leafy green vegetables and some vegetable oils. Table 2 shows the concentrations in some common foods (Booth et al., 1995; Shearer et al., 1996; Booth & Suttie, 1998).

Table 2. Phylloquinone content of common foods.

Table 2

Phylloquinone content of common foods.

The Total Diet Study of the US Food and Drug Administration is conducted periodically to monitor the safety and nutritional quality of the US food supply by assessing the levels of nutrients and contaminants in daily diets. It is based on the collection and analysis of 265 core foods. Intakes are estimated from the concentrations of individual nutrients and contaminants in the core foods and the mean consumption of the foods in each population group. The quantitative contributions of specific foods to the phylloquinone intake of the total population are presented in Table 3. Table 4 gives the estimated daily intake in 1990 for 14 categories of age and sex (Booth et al., 1996).

Table 3. Contribution of certain food groups to total adult intake (%) of phylloquinone in the USA, stratified by age and sex.

Table 3

Contribution of certain food groups to total adult intake (%) of phylloquinone in the USA, stratified by age and sex.

Table 4. Estimated and recommended mean dietary intakes of phylloquinone in the USA, stratified by age and sex.

Table 4

Estimated and recommended mean dietary intakes of phylloquinone in the USA, stratified by age and sex.

Phylloquinone has been determined by several analytical methods in human milk, in cow’s milk, in many brands of infant formula and in the oils that have been added to infant formulas for many years. Some of the concentrations found in each of these sources are presented in Table 1.

Menaquinones are synthesized by bacteria. They have a more restricted distribution in the diet than phylloquinone, and nutritionally significant amounts probably occur only in animal liver and some fermented foods, including cheese. Menaquinones are also synthesized by specific inhabitants of the human gut microflora. The major intestinal forms are MK-10 and MK-11 produced by Bacteroides, MK-8 by Enterobacteria, MK-7 by Veillonella genus and MK-6 by Eubacterium lentum (Shearer et al., 1996). The total concentration of menaquinones in human distal colonic contents is about 20 µg/g dry weight, with MK-10 predominating (Conly & Stein, 1992; Shearer, 1995). It seems likely that menaquinones synthesized by the gut microflora make a significant contribution to human tissue stores and are used by the hepatic vitamin K-dependent carboxylase, but the extent of this contribution remains uncertain (Shearer, 1995; Suttie, 1995).

1.5. Regulations and guidelines

Phylloquinone is listed (as phytomenadione or phytonadione) in the British, Chinese, Czech Republic, European, French, German, International, Japanese, Swiss and US pharmacopoeias (Royal Pharmaceutical Society of Great Britain, 1999; Swiss Pharmaceutical Society, 1999).

The Food and Drug Administration (1999) requires that all infant formulae sold in the USA contain a minimum of 4 µg/100 kcal (0.2 mg/kg) vitamin K; and that any vitamin K added should be in the form of phylloquinone.

Menadione is listed in the Austrian, Belgian, British, Dutch, European, French, German, International, Italian, Portuguese, Swiss and US pharmacopoieas, and menadione sodium bisulfite is listed in the Belgian, International, Swiss and US pharmacopoeias (Royal Pharmaceutical Society of Great Britain, 1999; Swiss Pharmaceutical Society, 1999). Menadiol sodium phosphate is listed in the British, Czech Republic and US pharmacopoeias (Swiss Pharmaceutical Society, 1999).

2. Studies of Cancer in Humans

The association between childhood cancer and vitamin K administered during the perinatal period with a view to preventing haemorrhagic disease of the newborn has been investigated in a number of studies (summarized in Table 5). The prophylactic use of vitamin K in newborns has varied with time, geographical location and among hospitals within countries. Some hospitals during some periods have had a selective policy based on the indications low birth weight, prematurity and operative delivery.

Table 5. Studies on childhood cancer and vitamin K administered during the perinatal period.

Table 5

Studies on childhood cancer and vitamin K administered during the perinatal period.

The hypothesis that vitamin K might be a risk factor for childhood cancer was generated on the basis of the results of a cohort study of 16 193 infants delivered in Great Britain in one week of April 1970, who were followed up at ages five and 10. The 33 cases included in the study were in patients who had died from cancer or were identified through cancer registration as having a cancer diagnosed before the age of 10. An unexpected statistically significant association was found between childhood cancer and administration of any drug during the first week of life (Golding et al., 1990), and 16 of the 18 patients who had received drugs during the first week of life had received vitamin K. Within the cohort, a comparison was made between the 33 cases and 99 controls matched with the cases for the age of the mother at the time of the birth of the child, parity, social class, marital status at delivery and whether the birth was single or multiple. Statistically significant associations were identified not only with drug administration during the first week of life, but also with antenatal X-rays, antenatal smoking, non-term delivery and use of pethidine or pethilorfan (a pethidine-containing drug) during labour. Only two of the 33 cases had fewer than two of these risk factors, whereas 45/99 (47%) of the controls had either no or only one risk factor. All but four of the mothers of the 16 cases who had received vitamin K had received pethidine or pethilorfan during labour. In a logistic regression analysis carried out on the whole cohort, in which social class was included with the other variables already mentioned, the relative risk associated with drug administration during the first week of life was 2.6 (95% confidence interval [CI], 1.3–5.2). [The Working Group noted that Cremophor EL was the only emulsifier used in Great Britain for vitamin K injection at the time (Draper & Stiller, 1992; Rennie & Kelsall, 1994; see Table 6.]

Table 6. Brands of vitamin K and vehicle used in different countries.

Table 6

Brands of vitamin K and vehicle used in different countries.

2.1. Case–control studies

In most of the case–control studies, the reference group comprised infants who had not received vitamin K and/or those who had received it orally. This combination is justified because the plasma concentrations after intramuscular administration are more than 10 times higher than those after oral administration (McNinch et al., 1985).

In a second study (Golding et al., 1992), 195 children with cancer diagnosed in the period 1971–91 who had been born at two major maternity hospitals in Bristol, England, in the period 1965–87 were compared with 558 controls identified from the delivery books of these hospitals. The cases were ascertained from the oncology register of the regional paediatric oncology unit and from the National Registry of Children’s Tumours. The basic method of control selection was to select every 300th birth in each year in each hospital. In view of the observation that the immediate effects of identical oral and intramuscular doses of vitamin K are different, the investigators sought to distinguish the effects of administration by the two routes. When the route of vitamin K administration was not recorded in the neonatal notes, a route was imputed on the basis of year of birth, the type of delivery and whether or not the infant was admitted to special care; the imputed route was identified in the absence of knowledge of case or control status. On the basis of 180 cases (92% of those for which notes were available) and 544 controls (98% of those for which notes were available), the relative risk (adjusted for hospital and year of delivery) for childhood cancer associated with intramuscular vitamin K was 2.2 (95% CI, 1.1–4.4) when compared with no vitamin K and 1.2 (95% CI, 0.5–2.7) for oral vitamin K. In view of the absence of an association with oral vitamin K in these data, the authors conducted a subsequent analysis in which the reference group was defined to include infants who had not received vitamin K or who had received it orally. The relative risk for leukaemia associated with intramuscular vitamin K was 2.7 (95% CI, 1.3–5.2) and that for other types of childhood cancer was 1.7 (95% CI, 1.0–2.8). Thus, there was no clear difference in the association by type of childhood cancer. When the analysis was confined to records in which the route was clearly stated, the odds ratio for all childhood cancer was 2.0 (95% CI, 1.2–3.3). These results could not be accounted for by other factors associated with the administration of intramuscular vitamin K, such as type of delivery or admission to a special care unit. Data were collected on 319 variables for all controls and for 111 cases of cancer ascertained from the oncology register of the regional paediatric oncology unit; these data were not obtained for the remaining 84 cancer cases. Of these variables, the presence of rubella antibody, resuscitation by intermittent positive pressure and paediatric estimate of gestation were statistically significant at the 1% level, which is what would be expected by chance. Adjustment for these and other variables reported to be associated with childhood cancer or known to be indicators for administering intramuscular vitamin K had little effect on the odds ratio for childhood cancer associated with vitamin K. Nineteen of the cases were diagnosed in the first year of life, and the possibility was considered that these cancers might have been present before the child was born and could therefore not have been initiated by an injection of vitamin K; however, the association persisted after exclusion of these 19 cases from the analysis. When the analysis was restricted to subjects who would have been followed for at least 10 years, by considering only those born in the period 1971–80, the relative risk for all childhood cancer associated with intramuscular vitamin K was 1.9 (95% CI, 1.1–3.4), similar to that assessed for all subjects. [The Working Group noted, as acknowledged by the authors, a large number of instances in which the information on potentially confounding variables was not available, for example on smoking in pregnancy. Medical records are not necessarily reliable sources of information about pregnancy and childbirth (Hewson & Bennett, 1987; Oakley et al., 1990), and this, together with the fact that potential confounding was assessed only for a subset of cases, constitutes a limitation of the study. The relationship between the type of delivery and intramuscular administration of vitamin K differed markedly between the two maternity hospitals in Bristol in which the case and control subjects in the study had been born (Carstensen, 1992; Draper & Stiller, 1992). The association with childhood cancer is largely accounted for by data from one of the hospitals in which virtually all of the control infants who received intramuscular vitamin K had been born by an assisted delivery. This raises the issue as to whether bias arose in control selection in that hospital.]

A study in the USA was reported by Klebanoff et al. (1993) which was based on follow-up to the age of seven or eight years of 54 795 liveborn children of women enrolled between 1959 and 1966 in 12 centres contributing to the National Collaborative Perinatal Project. Neonates whose cancer was diagnosed or strongly suspected during the first day of life were excluded because vitamin K could not have been a factor in those cases. Vitamin K was administered in the delivery room or the nursery, and information about the administration was recorded with other events during and after delivery by observers who were not involved in the clinical care of the mother or the infant. Cancer was diagnosed in 48 of 54 795 liveborn children after the first day of life. For each case, five controls were selected and matched with the index case on length of follow-up. In spite of the prospective recording by the observers, the data on vitamin K administration were not recorded unambiguously for 43 infants; a review of hospital records without knowledge of case or control status resulted in data for 25 (58%) of these. The exposure status was unknown for four case children. The relative risk for all childhood cancer associated with vitamin K was 0.84 (95% CI, 0.41–1.7), and that for leukaemia was 0.47 (95% CI, 0.14–1.6; based on 15 cases). In the USA, only two brands, Aquamephyton and Konakion, have been approved for use (see Table 6). Konakion in the USA contains polysorbate-80 rather than Cremophor EL as an emulsifier and phenol as an antrimicrobial agent. In the study of Klebanoff et al. (1993), the relative risk for total childhood cancer associated with the two brands together was 0.6, whereas that for children who had received the phenol-containing preparation alone was 0.7. In this study, only one child had received vitamin K orally.

von Kries et al. (1996) carried out a case–control study of children born in 162 obstetric hospitals in Lower Saxony (Germany) during the period 1975–93 when only one vitamin K preparation, Konakion, the same as that used in the United Kingdom, was licensed for neonatal vitamin K prophylaxis. Of a total of 218 children with leukaemia identified as eligible, information on vitamin K prophylaxis was obtained for 136 (62%). For each leukaemia case, one control was selected from the municipality where the patient lived at the time of diagnosis (local control), and a second one (state control) from a municipality selected at random in Lower Saxony by means of a population-weighted sampling scheme. These controls were matched with cases by sex and date of birth. Case and control families were contacted initially by being sent a questionnaire. If a control family refused to collaborate in the study or did not return the questionnaire within three months, another control family was invited; control families that returned the questionnaire after more than three months were also included. Thus, a total of 305 local and 308 state controls were invited to participate. Information on vitamin K prophylaxis was obtained for 174 (57%) of the local controls and 160 (52%) of the state controls. As the study was performed as part of a population-based case–control study to explore possible causes of childhood leukaemia in Lower Saxony, a third control group for the leukaemia study was identified which comprised cases of brain tumours, nephroblastoma, neuroblastoma and rhabdomyosarcoma. No population-based controls were selected for these cases, but they were used as additional cases in the study of vitamin K. Of a total of 246 potentially eligible cases of this type, information on vitamin K prophylaxis was obtained for 136 (55%). Data on vitamin K prophylaxis were abstracted from the birth report with no knowledge of the case or control status of each child. Information on the dose and route of vitamin K prophylaxis was obtained from the birth record or in the delivery book for 72% of the 272 cases of leukaemia and other cancers and 64% of the 334 controls. When this information was not available, the index child was assumed to have had the same exposure to vitamin K as the child nearest to the index infant in the delivery book with the same route of delivery and same perinatal morbidity (nine cases and six controls). When this could not be established, staff who worked in the delivery unit at the time when the index child was born were asked what kind of vitamin K prophylaxis the index infant would have received, given the birth weight and route of delivery (63 cases and 109 controls). Finally, similar information was sought from medical staff who did not work in the delivery unit at the time the index child was born (four cases and four controls). In the comparison with local controls (n = 107), the risk for leukaemia (n = 107) associated with intramuscular or subcutaneous administration of vitamin K relative to that for oral or no vitamin K prophylaxis was 1.2 (95% CI, 0.68–2.25). In the comparison with state controls (n = 160; leukaemia cases = 136), the relative risk was 0.82 (95% CI, 0.50–1.4). When the control groups were pooled (n = 334), the relative risk was close to unity (136 leukaemia cases), and the relative risk for brain tumours, nephroblastoma, neuroblastoma and rhabdomyosarcoma combined (n = 136) associated with vitamin K prophylaxis was 1.2 (95% CI, 0.77–1.8). When the analyses were repeated for subjects for whom vitamin K prophylaxis had been documented in birth records or delivery books, the results were almost unchanged, except in the comparison of leukaemia cases with local controls, which gave a relative risk of 2.0 (95% CI, 0.69–6.0). When the analyses were repeated for parenteral prophylaxis versus no prophylaxis, most of the relative risks were slightly decreased. The risk of the subgroup of cases of leukaemia in children aged 1–6 years was analysed as this was considered to be a relatively homogeneous subgroup, most of the cases having common acute lymphoblastic leukaemia. [The Working Group noted that it is not clear whether the decision to make this subgroup analysis was specified in the original study protocol or was made post hoc.] The risk relative to both control groups combined was 1.2 (95% CI, 0.69–2.15), in the comparison with state controls it was 0.99 (95% CI, 0.52–1.9) and in the comparison with local controls it was 2.3 (95% CI, 0.94–5.5). There was no difference between cases and controls in the source of information on vitamin K prophylaxis. The increased relative risk in the comparison with local controls could not be explained by any of the potential confounders. It would be expected that the policy of administration of vitamin K would be more likely to be similar for cases and local controls than for cases and state controls. Therefore, the relative risk would be expected to be closer to unity in the comparison between cases and local controls than in the other comparison, whereas the opposite was observed. The non-significantly increased risk relative to local controls may be a chance result in subgroup analysis with multiple testing, as acknowledged by the authors.

In a case–control study of childhood leukaemia based on births in three hospitals in England (Cambridge, Oxford and Reading), no association with intramuscular vitamin K, either as determined from hospital records (91 cases, 171 controls) or as imputed from hospital policy (132 cases, 264 controls), was found. In addition, no association was found specifically for acute lymphoblastic leukaemia (Ansell et al. 1996). Subsequently, Roman et al. (1997) reported a more detailed analysis of data on leukaemia and non-Hodgkin lymphoma diagnosed before the age of 30 years in subjects whose obstetric records were stored in the same three hospitals. Ninety-two per cent (132/143) of the cases of leukaemia were diagnosed at age 14 or less; these cases and their controls were included in the report of Ansell et al. (1996). There was no association between leukaemia and intramuscular vitamin K administration either recorded in the notes (relative risk, 1.2; 95% CI, 0.7–2.1) or imputed from information about hospital policy (relative risk, 1.2; 95% CI, 0.5–2.4). In view of the finding of von Kries et al. (1996), acute lymphoblastic leukaemia diagnosed between the ages of 1–6 years was considered; the relative risk associated with recorded administration (based on hospital notes) was 0.6 (95% CI, 0.3–1.4), and that based on hospital policy was again 0.6 (95% CI, 0.2–1.7).

Parker et al. (1998) identified 1432 children born in northern England between 1960 and 1991 from the regional Children’s Malignant Disease Registry, in whom cancer was diagnosed in 1968–92 when they were aged between three months and 14 years while still resident in the region. The birth records of 701 of these children could not be traced, usually because the maternity unit had retained only its most recent records or because the unit had closed and the records could not be located. Thirty children who had been given vitamin K orally at birth and 16 cases in multiple births were excluded. The controls were selected by taking the fourth, eighth and 12th birth before and after the index birth from birth or admission registers for the hospital of birth of the index child. Towards the end of the study, the number of controls per case was reduced from six to three because of time constraints. When the birth notes for control children could not be located, or when the child selected was found to be on the Malignant Disease Register, the next possible control was selected. The fact of intramuscular administration of vitamin K or non-administration of vitamin K was recorded in the maternity unit records for 438 of 685 cases (case notes). [The Working Group noted that the corresponding proportion for controls was not specified.] There was no association between intramuscular vitamin K administration and either all cancers or all cancers other than acute lymphoblastic leukaemia. The relative risk for acute lymphoblastic leukaemia associated with vitamin K administration based on case notes was 1.4 (95% CI, 0.71–1.7; 132 cases). Two secondary analyses were conducted to consider cases typical of the peak incidence of leukaemia in early childhood. When the 51 children in the case note analysis who had T-cell leukaemia or for whom subtype characterization was not available were excluded, the relative risk for the 81 cases of non-T-cell lymphoblastic leukaemia was 1.8 (95% CI, 0.82–3.9). In an analysis of 94 children aged 1–6 years at diagnosis, the relative risk was 2.3 (95% CI, 0.98–5.2). In all of these analyses, adjusted relative risks were calculated separately for the specified potential confounding factors—sex, gestation, birth weight, opiates during labour, assisted delivery, signs of asphyxia at birth, admission to special care or neonatal blood transfusions. Except for adjustment for assisted delivery, admission to special care or opiate exposure in labour, none of these changed any of the relative risks by more than 10%. Adjustment for assisted delivery or admission to special care caused a larger rise in the relative risk. The relative risk for acute lymphoblastic leukaemia diagnosed at ages 1–6 was 2.4 (95% CI, 1.0–5.7) after adjustment for exposure to opiates and 3.6 (95% CI, 1.3–9.7) after adjustment for assisted delivery based on case note analysis. As in many of the other studies, information on hospital policy was obtained in order to impute exposure when this was unclear from medical records. This information was obtained by a research midwife and neonatal staff in each unit in the region and by a paediatrician from current and recently retired medical staff, and this independently obtained information was then cross-validated. When inconsistencies were identified, the case notes were sampled to determine what policy had actually been followed. This enabled a further 226 cases to be included at the analysis; 21 cases were excluded because the policy of the local unit could not be ascertained. The relative risks were similar to but somewhat lower than those in the analysis based exclusively on subjects for whom data on vitamin K exposure was obtained only from medical records. [The Working Group noted that it was unclear which hypotheses about subgroups had been pre-specified. Bias may have arisen from the fact that while a large proportion of cases had to be excluded there was a mechanism for adding controls when a control record was unobtainable. Availability of records might have associations with both perinatal health problems and subsequent development of childhood cancer.]

McKinney et al. (1998) carried out a case–control study on childhood cancer in Scotland using data abstracted from 76 hospital records. A total of 500 cases of cancer diagnosed in children aged 0–14 years during the period 1991–94 while resident in Scotland were identified. Controls matched on age, sex and health board of residence were randomly selected from among all eligible children registered for primary care within each health board. A total of 1338 eligible controls was identified. A total of 460 mothers of cases (92%) and 861 mothers of controls (64%) were interviewed, and medical notes were abstracted for 440 cases and 802 controls. The data set for statistical analysis was restricted to matched sets, and information was lost for 23 cases without matched controls and 25 controls without a matched case. Therefore, 417 cases and 777 controls were included in the matched case–control analysis. Vitamin K was recorded as given or definitely not given only when this was mentioned in the notes. Similarly, the route of administration was classified as intramuscular, oral or not recorded. None of the relative risks reported for leukaemias, acute lymphoblastic leukaemia, lymphomas, central nervous system tumours or other solid tumours, either crude or adjusted for social class and type of delivery, was statistically significantly different from unity. The adjusted relative risk for leukaemia associated with vitamin K given intramuscularly (recorded) in the neonatal period was 1.2 (95% CI, 0.77–2.0) and that for acute lymphoblastic leukaemia was 1.2 (95% CI, 0.70–2.0). In view of the findings of Parker et al. (1998, see above), the subset of acute lymphoblastic leukaemia diagnosed in children aged 1–6 years (90 cases, 174 controls) was also analysed, and the adjusted relative risk was found to be 1.2 (95% CI, 0.62–2.2). As nothing about vitamin K had been written in the medical records for a substantial proportion of children (37% of cases and 35% of controls), the authors also sought to impute exposure on the basis of hospital policies. Information on the vitamin K policies of hospitals in which over 500 infants were delivered annually was validated by abstraction of a sample of medical records and through consultations with hospital pharmacies and senior labour room midwives. For 100 (24%) cases and 191 (25%) controls, no hospital policy was available for any imputation. The relative risks for the specific diagnostic categories associated with intramuscular vitamin K administration in the neonatal period either as recorded in medical records or imputed from hospital policy were very similar to those calculated for subjects for whom only data from medical records were included. The adjusted relative risk for leukaemia was 1.3 (95% CI, 0.78–2.1), that for acute lymphoblastic leukaemia was 1.1 (95% CI, 0.65–1.9) and that for acute lymphoblastic leukaemia in children aged 1–6 years was 1.3 (95% CI, 0.70–2.5). Very few subjects were recorded as having or imputed to have been given vitamin K orally in the neonatal period (12 cases, 2.9%; and 33 controls, 4.3%).

Passmore et al. (1998a) identified cases of childhood cancer diagnosed at ages up to 14 years in persons who were resident in Great Britain and had been born in 16 hospitals with large maternity units in 1968 or later and diagnosed by the end of 1986 from the National Registry of Childhood Tumours (excluding retinoblastoma, Down syndrome or neurofibromatosis). The 16 hospitals were selected on the basis of a survey which showed that they had a selective policy for the use of vitamin K prophylaxis. Of 1092 cases initially identified as born in these hospitals, 523 were born in the years for which a policy was known and for whom the medical records were found. Four controls matched on sex, month of birth and hospital of birth were selected randomly from these registers. Medical records departments were asked to locate the records for each case and for one control. Initially, two out of each of the four potentially eligible controls were selected randomly for location by the medical records department. If the records department was unable to locate the notes of either of these, details were supplied of the other two. Controls with illegible records, twins, stillbirths and neonatal deaths were excluded. In addition, infants with severe neural tube defects or a birth weight of less than 1000 g were excluded, as they were unlikely to have survived to the age at which the case patient developed cancer. For these, an alternative control was selected by using the next suitable birth in the hospital birth register. [The numbers of control replacements were not specified.] A second group of cases from the same period was chosen from records of the National Registry of Childhood Tumours in order to identify cases of cancer among children included in a survey of more than 100 000 births in South Glamorgan, Wales. For each case, two controls matched for sex, month of birth and hospital were selected, applying the same set of exclusions. Medical records were sought for all cases and controls, and information on vitamin K administration taken from these records was supplemented by data from the birth survey, which was available for most but not all of the period of study. This added three further hospitals to the study, all of which had selective policies of vitamin K administration, and 74 cases. In the combined data (16 maternity units in England and Wales and the three hospitals included in the survey in South Glamorgan), the relative risk for childhood cancer of all types associated with intramuscular vitamin K administration was 1.4 (95% CI, 1.0–2.1). In the data for the 16 maternity units in England and Wales, the relative risk was 1.2 (95% CI, 0.77–1.9), while in the data from South Glamorgan, the relative risk was 2.1 (95% CI, 1.1–4.1). For the combined data and for the data from South Glamorgan, mode of delivery (forceps, vacuum extraction, breech or caesarean) was a statistically significant confounding variable, and adjustment for this reduced the relative risks to 1.1 for the combined data and 1.3 for the South Glamorgan data. In the combined data, the relative risk for leukaemia was 1.5 (95% CI, 0.82–2.85), that for acute lymphoblastic leukaemia was 1.7 (95% CI, 0.89–3.3) and that for acute lymphoblastic leukaemia diagnosed at ages 1–5 years was 1.0 (95% CI, 0.48–2.2). Again, adjustment for mode of delivery reduced the relative risks. [The Working Group noted that the substantially lower relative risk for the 1–5 year-old group than for all ages combined implies that the effect for children of other ages is higher than that for this group, in contrast to the observations of von Kries et al. (1996) and Parker et al. (1998).] The relative risk for nonleukaemia cancers was 1.4 (95% CI, 0.88–2.2) in the combined data and 2.4 (95% CI, 1.1–5.4) in the data from South Glamorgan. In the South Glamorgan data, none of the potential confounders that were adjusted for reduced the magnitude of the relative risk. [The Working Group noted that in the absence of an effect in the data from the 16 maternity units in England and Wales, the South Glamorgan finding may reflect an unidentified bias or be a chance finding.]

[The Working Group noted that in the subgroup analyses of acute lymphoblastic leukaemia diagnosed at 1–6 years carried out by Parker et al. (1998) and 1–5 years by Passmore et al. (1998a), adjustment for mode of delivery had contrasting effects. In the study of Passmore et al. it attenuated the relative risk associated with vitamin K, while in the study of Parker et al. the relative risk was increased.]

2.2. Ecological studies

These studies are summarized in Table 7.

Table 7. Ecological studies on childhood cancer and vitamin K administered intramuscularly during the perinatal period as Konakiana.

Table 7

Ecological studies on childhood cancer and vitamin K administered intramuscularly during the perinatal period as Konakiana.

Ekelund et al. (1993) investigated the association between childhood cancer and intramuscular administration of vitamin K in a study in Sweden based on linkage of the medical birth registry to the national cancer registry. The study was restricted to fullterm infants (gestation, 37–42 weeks) who had survived and who were born in 1973–89 after a delivery without use of forceps or vacuum extraction. The infants were followed up to 1 January 1992. Cancers diagnosed within 30 days of birth were regarded as congenital and were excluded from the analysis. Routines for administration of vitamin K were obtained from all 95 maternity hospitals and validated for a subset of 102 children with cancer and 100 control children randomly selected from among those who, according to the information on routine exposure, received intramuscular vitamin K, and 94 children with cancer and 100 control children from among those who should have received oral vitamin K. The doses of vitamin K given in Sweden were similar to those given in the United Kingdom, and the same preparation was used (phylloquinone, Konakion, see Table 6). When the method of administration of vitamin K was recorded, it agreed with the stated routine method of administration in 92% of the 235 cases for which individual information could be found. The relative risk for all childhood cancer associated with a hospital policy of intramuscular administration of vitamin K as compared with oral administration was 1.0 (95% CI, 0.88–1.2, after stratification for year of birth). The relative risk for leukaemia was 0.90 (95% CI, 0.70–1.2).

Olsen et al. (1994) compared the cumulative risk of childhood cancer among children aged 1–15 years who were born during the period 1945–54 (n = 835 430), in which no vitamin K was administered, those aged 1–15 years born during the period 1960–69 (n = 797 472), in which pregnant women received oral vitamin K, and those aged 1–13 years born during the period 1975–84 (n = 586 378), in which virtually all newborns received vitamin K intramuscularly. There was a small increase in risk for all tumour types combined, due mainly to lymphoma in boys and neuroblastoma in boys and girls. There was no trend for childhood leukaemia. The preparation was the same as that used in the United Kingdom (Draper & McNinch, 1994).

In addition to the case–control study in northern England described above, Parker et al. (1998) compared the incidence of acute lymphoblastic leukaemia diagnosed in children aged up to 14 years who were born in hospital units in which all infants received vitamin K, with those born in units where less than a third received this prophylaxis. As described above, information on hospital policy was obtained separately and independently by two people and then cross-validated. In units with a policy of selective prophylaxis, less than 30% of infants received intramuscular vitamin K at birth, while in units offering universal prophylaxis, sampling of case notes showed that more than 95% of babies received vitamin K. The risk for acute lymphoblastic leukaemia in children born in hospitals with a policy of universal prophylaxis relative to those born in hospitals with a policy of selective prophylaxis was 0.95 (95% CI, 0.78–1.2). The relative risk of the subgroup diagnosed at 1–6 years was 1.05 (95% CI, 0.82–1.35). [The Working Group noted that the cases included in this analysis overlapped with those included in the case–control study, so that the results are not independent].

Passmore et al. (1998b) carried out a similar comparison of cancers of all types other than retinoblastoma or associated with Down syndrome or neurofibromatosis diagnosed in children aged 1–14 years who were born in 94 hospital units in Great Britain. Information on hospital policy for neonatal vitamin K was obtained during the case–control studies of Passmore et. al. (1998a) and Ansell et al. (1996), described above, for 30 hospitals in Scotland from members of the Scottish Neonatal Network and from paediatricians for 41 of a further 80 hospitals in England and Wales in which more than 25 children who subsequently developed cancer had been born in the period 1968–85. The observed numbers of cases in hospitals with universal and selective policies were compared with the numbers expected on the basis of national rates. Separate analyses were carried out for births in hospitals that followed one policy throughout the period of study and births in hospitals in which the policy changed during the period of study. A large number of observed:expected ratios were calculated. The ratio for all cancers was 0.97, that for leukaemia at 1–14 years was 1.03, and that for acute lymphoblastic leukaemia at 1–5 years was 1.01 for hospitals with a consistent, non-selective policy. The ratio tended to be smaller in hospitals with a selective policy than in those offering universal prophylaxis. The only statistically significant (p < 0.05, two-tailed test) departure from unity indicated a lower risk for cancer other than leukaemia among children born in hospitals offering universal prophylaxis that those born in hospitals consistently offering selective prophylaxis in Scotland. [The Working Group noted that the cases included in this analysis overlapped with those in the case–control studies of Parker et al. (1998) and Ansell et al. (1996), so that the results are not independent.]

3. Studies of Cancer in Experimental Animals

No reports of studies specifically designed to investigate the carcinogenicity of vitamin K substances were available to the Working Group. One study on the initiating effects of menadione in an assay of liver foci in rats was available (Denda et al., 1991) but could not be evaluated owing to methodological limitations.

4. Other Data Relevant to an Evaluation of Carcinogenicity and its Mechanisms

The studies summarized in this section should be considered in the light of the differences between naturally occurring forms of vitamin K that have a lipophilic side-chain at the 3-position of the 2-methyl-1,4-naphthoquinone (menadione) ring structure (phylloquinone and menaquinones) and the synthetic forms which lack this side-chain (menadione and its water-soluble derivatives). Lack of this side-chain results in profound differences in the absorption, tissue distribution and metabolism of natural K vitamins. Importantly, the lack of a lipophilic side-chain is the reason for the increased chemical reactivity and greater toxicity of menadione when compared with phylloquinone and menaquinones. In the strict sense, menadione is a provitamin K, because it is biologically active for the synthesis of vitamin K-dependent proteins only after conversion to the naturally occurring menaquinone-4 (four prenyl units) in vivo.

4.1. Absorption, distribution, metabolism and excretion

4.1.1. Humans
(a) Intestinal absorption and plasma transport in adults

The major dietary form of vitamin K is phylloquinone (Shearer et al., 1996). It is absorbed chemically unchanged from the proximal intestine after solubilization into mixed micelles composed of bile salts and the products of pancreatic lipolysis. In healthy adults, the efficiency of absorption of phylloquinone in its free form is about 80% (Shearer et al., 1974), but the efficiency of absorption from green leafy vegetables such as spinach is < 10% (Gijsbers et al., 1996).

Within the intestinal mucosa, phylloquinone is incorporated into chylomicrons, is secreted into the lymph and enters the blood via the lacteals (Shearer et al., 1970, 1974). After a phylloquinone-containing meal, the plasma concentration peaks between 3 and 6 h (Shearer et al., 1970; Lamon-Fava et al., 1998). Once in the circulation, phylloquinone is rapidly cleared at a rate consistent with its continuing association with chylomicrons and the chylomicron remnants that are produced by lipoprotein lipase hydrolysis at the surface of capillary endothelial cells. During the postprandial phase and after an overnight fast, more than half of the circulating phylloquinone is associated with triglyceride-rich lipoproteins, and the remainder is carried by low-density and high-density lipoproteins (Kohlmeier et al., 1996; Lamon-Fava et al., 1998). Although phylloquinone is the major circulating form of vitamin K, menaquinone-7 is present in plasma at lower concentrations and has a similar lipoprotein distribution to phylloquinone. While phylloquinone in blood is derived exclusively from the diet, it is not known what proportion of circulating menaquinones such as menaquinone-7 derives from the diet or the intestinal flora (Shearer et al., 1996).

(b) Plasma pharmacokinetics of phylloquinone in adults

The plasma clearance of an intravenous dose of 1 mg [3H]phylloquinone during the first 6 h resolved approximately into two exponential functions, the first with a half-time of 20–24 min and the second with a half-time of 121–150 min (Shearer et al., 1972). The curves for clearance up to 12 h after an intravenous injection of a 10-mg dose of phylloquinone (Konakion MM) were similar to those after 1 mg and were consistent with a two-compartment (sometimes three-compartment) model in which the log-linear terminal phase over 3–12 h had a half-time of about 3 h (Soedirman et al., 1996). A gradual slowing of the clearance rate was seen after the first 6 h (Shearer et al., 1972, 1974), as was also found in a study of the clearance of pharmacological doses of 10–60 mg by Øie et al. (1988), who reported that the log-linear terminal elimination phase was not reached before 8–12 h and that the average half-time was 14 h (range, 8–22 h). This slowing of the clearance rate may be explained by the complexity of the plasma transport of phylloquinone, in which the proportion of phylloquinone associated with low-density and high-density lipoproteins increases progressively (Lamon-Fava et al., 1998).

The plasma disposition of oral doses of 5–60 mg phylloquinone (Konakion or AquaMephyton) is similar to that found after a more physiological dose (≤ 1 mg), with peak plasma concentrations at 4–6 h followed by a rapid clearance phase (Shearer et al., 1974; Park et al., 1984; Øie et al., 1988; Hagstrom et al., 1995). After an oral dose of 10 or 50 mg Konakion, the plasma concentration declined from the peak absorptive level at a similar log-linear rate as that seen after intravenous administration, with a terminal half-time of about 2 h for measurements up to 9–12 h (Park et al., 1984). The absorption of oral preparations of phylloquinone shows inter- and intra-individual variation and, for doses of Konakion ranging from 10 to 60 mg, the bioavailability was 10–63% (Park et al., 1984) and 3.5–60% (Øie et al., 1988).

The pharmacokinetics of phylloquinone after an intramuscular dose is completely different, showing sustained, slow release from the muscle site over many hours and marked inter-individual variation (Hagstrom et al., 1995; Soedirman et al., 1996). The pharmacokinetics may also be influenced by the solubilizing agent. The systemic availability of intramuscularly injected Konakion MM, which is a mixed-micellar solution of phylloquinone in natural solubilizers, the bile acid glycocholic acid and the phospholipid lecithin (Schubiger et al., 1997; see Table 6), was irregular and < 65% in 20% of subjects (Soedirman et al., 1996). After intramuscular injection of phylloquinone (AquaMephyton R), most of the substance was carried by low-density and high-density lipoproteins instead of by triglyceride-rich (very-low-density) lipoproteins as found after oral administration (Hagstrom et al., 1995).

(c) Plasma pharmacokinetics of phylloquinone in neonates

The pharmacokinetics of phylloquinone during the early clearance phase up to 6 h in neonates (of low birth weight) after intravenous injection was very similar to that of adults (Shearer et al., 1972), declining bi-exponentially with median half-times of 23 and 109 min (Sann et al., 1985).

An early study of the plasma disposition of 1 mg Konakion given orally or intramuscularly at birth showed wide inter-individual differences during the first 24 h, especially after oral administration (McNinch et al., 1985). The peak plasma concentration after an oral dose occurred after 4 h; the median concentration was 73 ng/mL, which fell to 23 ng/mL after 24 h. The plasma concentration after administration of 1 mg of Konakion intramuscularly exceeded those after oral administration at all times, and after 24 h the median was 444 ng/mL. Physiologically, these concentrations compare with adult endogenous levels of about 0.5 ng/mL (Shearer, 1992).

In a comparison of the plasma concentrations of Konakion and Konakion MM in exclusively breast-fed infants at 24 h and 4 and 24 days after a single oral dose of 2 mg at birth (Schubiger et al., 1997), the mixed-micellar Konakion MM preparation resulted in higher median concentrations at all times, suggesting greater bioavailability. The largest difference was seen after four days, with median concentrations of 41 ng/mL Konakion MM and 12 ng/mL Konakion. By 24 days, the concentrations in both groups were mainly within the adult physiological range (0.3–0.4 ng/mL). An earlier study by the same group (Schubiger et al., 1993) had shown that a single oral dose of 3 mg Konakion MM resulted in higher plasma concentrations than a single dose of 1.5 mg of the same preparation given intramuscularly after four days. In this study, however, the plasma concentrations after 24 days were significantly higher after intramuscular injection, consistent with the hypothesis of the depot effect of intramuscular phylloquinone (Loughnan & McDougall, 1996; see also section 4.1.1(f)).

Stoeckel et al. (1996) pointed out that the terminal elimination plasma half-time of phylloquinone in neonates is probably longer than that in adults. They calculated from published studies that a realistic estimate of the terminal plasma half-time in neonates was 26–193 h (median, 76 h), as compared with 8–22 h (median, 14 h) in adults after intravenous administration (Øie et al., 1988). This longer terminal half-time may reflect the poorly developed organ systems of neonates and a reduced capacity to metabolize and excrete vitamin K (Stoeckel et al., 1996).

(d) Plasma pharmacokinetics of menaquinone-4

Oral preparations of menaquinone-4 are used in Japan for the prophylaxis of vitamin K deficiency bleeding. The plasma profile of an oral dose of this preparation in five-day-old infants appeared to be similar to that of phylloquinone; after a 4-mg dose, a peak concentration of about 100 ng/mL was achieved after 3–4 h, before declining to about 30 ng/mL by 12 h (Shinzawa et al., 1989). The half-time of menaquinone-4 was not calculated.

(e) Adult tissue reserves and distribution of vitamin K

Dietary vitamin K is delivered to the liver and possibly other tissues, including bone marrow, in the form of chylomicron remnants (Kohlmeier et al., 1996). The liver has often been assumed to be a major depot for vitamin K because it is the site of synthesis of the vitamin K-dependent coagulation proteins. Measurements of phylloquinone in livers obtained at autopsy from 32 adults in the United Kingdom revealed hepatic concentrations ranging from 1.1 to 21 ng/g wet tissue [2.4–47 pmol/g], with a median concentration of 5.5 ng/g [12 pmol/g]. The corresponding total liver stores of phylloquinone were 1.7–38 µg [3.8–85 pmol/g], with a median total store of 7.8 µg [17 pmol/g] (Shearer et al., 1988). Similar hepatic concentrations of phylloquinone were found in a smaller number of analyses of post-mortem samples from adults in Japan (10 ng/g) (Uchida & Komeno, 1988) and in The Netherlands (11 ng/g) (Thijssen & Drittij-Reijnders, 1996). The limited ability of the liver to store vitamin K is illustrated by the observation that the phylloquinone reserves are about 40 000-fold lower than those of vitamin A despite a daily dietary intake of vitamin K (∼100 µg) which is only about 10-fold lower than that of vitamin A (∼1000 µg). The distribution of the various forms of vitamin K in the liver is quite different from that in plasma in that the major transport form, phylloquinone, represents the minority of total hepatic stores (about 10%); the remainder comprises bacterial menaquinones, mainly menaquinones-6–13 (Shearer et al., 1988; Shearer, 1992; Shearer et al., 1996). The pattern of individual menaquinones in the liver varies considerably between individuals (Shearer et al., 1988; Uchida & Komeno, 1988; Thijssen & Drittij-Reijnders, 1996), perhaps reflecting their origin from the intestinal microflora (Shearer et al., 1996). This proposal is supported by the finding that two menaquinones, -10 and -11, which are major forms in most liver samples (Uchida & Komeno, 1988; Thijssen & Drittij-Reijnders, 1996), are known to be synthesized by Bacteroides species which are predominant members of the human intestinal flora (Conly & Stein, 1992); yet menaquinone-10 and menaquinone-11 do not make appreciable contributions to normal diets (Shearer et al., 1996).

Phylloquinone is also present in other human tissues. The concentration in the heart (∼5 ng/g) [∼10 pmol/g] is comparable to those in the liver, and even higher concentrations (∼13 ng/g) [∼25 pmol/g] are found in the pancreas, but lower concentrations (< 1 ng/g) [< 2 pmol/g] were detected in brain, kidney and lung. These tissues do not appear to contain appreciable concentrations of menaquinones except for the short-chain menaquinone-4. Particularly high concentrations of menaquinone-4 relative to phylloquinone are present in the kidney, brain and pancreas. Although these and other tissues contain the enzymes of the vitamin K epoxide cycle (see Figure 1) and carry out vitamin K-dependent carboxylation of protein precursors, this would not appear to account for the tissue-specific accumulation of menaquinone-4 and may suggest a hitherto unrecognized physiological role for menaquinone-4 in certain tissues (Shearer, 1992; Thijssen & Drittij-Reijnders, 1996). Indeed, menaquinone-4 may arise by tissue synthesis from phylloquinone itself (Davidson et al., 1998).

Figure 1

Figure 1

Cyclic metabolism of vitamin K for conversion of glutamate (Glu) residues to γ-carboxy glutamate (Gla) residues in vitamin K-dependent proteins

Osteocalcin is a major vitamin K-dependent bone protein synthesized by osteoblasts and therefore requires a source of vitamin K for γ-glutamyl carboxylation. Both trabecular and cortical bone contain ample reserves of vitamin K, with phylloquinone predominating and smaller amounts of shorter-chain menaquinones (Hodges et al., 1993; Shearer, 1997). With the absence of the typical hepatic forms menaquinones-10–13, the vitamin K content of bone resembles that of other extrahepatic tissues.

(f) Tissue stores and blood concentrations in neonates and infants

Information on liver stores (the site of synthesis of vitamin K-dependent clotting proteins) in infants and their response to vitamin K prophylaxis is limited (Shearer et al., 1988; Guillaumont et al., 1993). The endogenous stores of vitamin K in the liver of the newborn differ both quantitatively and qualitatively from those of adults because the concentrations and total reserves of phylloquinone are lower than those of adults (Shearer et al., 1988) and because bacterial menaquinones are undetectable (Shearer et al., 1988; Guillaumont et al., 1993). The endogenous hepatic concentrations of phylloquinone ranged from 0.3 to 6.0 ng/g (median, 1.4 ng/g) in preterm infants and from 0.1 to 8.8 ng/g (median, 1.0 ng/g) in term infants. The median hepatic concentration of 1 ng/g in term infants is equivalent to a total liver pool of about 0.1 µg phylloquinone, whereas the concentration is 5.5 ng/g and the pool 7.8 µg in adult liver. In infants who had received 0.5 or 1 mg phylloquinone at birth by intramuscular injection, these liver reserves were raised by some two to three orders of magnitude within 24 h. Hepatic phylloquinone concentrations may remain elevated for several weeks after injection: in two infants known to have received 1 mg phylloquinone by the intramuscular route and who survived 13 and 28 days, the total hepatic stores were 24 and 15 µg, respectively (Shearer et al., 1988). Guillaumont et al. (1993) measured hepatic concentrations in post-mortem liver samples obtained within the first 48 h of death from infants who had received 2 mg phylloquinone intravenously or orally (in some cases combined with extra intravenous or oral doses of 1, 5 or 10 mg). In three newborns who survived < 24 h, the hepatic concentrations of phylloquinone ranged from 63 to 94 µg/g (total liver stores, 2800–7300 µg), which were four orders of magnitude higher than the endogenous concentrations of 0.002–0.008 µg/g (total liver stores, 0.1–0.9 µg). Between 24 and 48 h, the hepatic concentrations in 10 infants had fallen to a median of 8.4 µg/g (total liver stores, 550 µg), and in one infant who survived for five days it was 2.9 µg/g (110 µg). The quite rapid fall in hepatic stores presumably reflects the relatively rapid metabolism and excretion of vitamin K via the urine and bile (Shearer et al., 1974). The lower hepatic concentration after intramuscular injection (Shearer et al., 1988) compared with intravenous injection (Guillaumont et al., 1993) is consistent with the idea that phylloquinone injected intramuscularly is released relatively slowly from the injection site (Loughnan & McDougall, 1996).

The reduced hepatic reserves of vitamin K in the human neonate are best explained by the existence of a barrier to placental uptake or transfer. This suggestion was originally made on the basis of the large concentration gradient of physiological concentrations of phylloquinone between maternal and cord blood plasma and the inefficient maternal–fetal transfer of pharmacological doses administered as an intravenous injection to the mother just before delivery (Shearer et al., 1982). The poor placental transport of phylloquinone has been confirmed by others (Mandelbrot et al., 1988; Yang et al., 1989). There is now general agreement that the cord plasma concentration of phylloquinone is < 50 pg/mL [110 pmol/L] and that the average maternal–fetal concentration gradient is within the range 20:1 to 40:1 (Shearer, 1992).

Few longitudinal studies have been conducted of plasma concentrations in infants who were not given vitamin K prophylaxis. In one such study, cord plasma concentrations were compared for breast-fed and formula-fed infants and in blood on days 3, 7 and 28 after birth (Pietersma-de Bruyn et al., 1990). In entirely breast-fed infants, the blood concentration rose from undetectable (< 20 pg/mL) at birth to mean values of 0.76, 0.49 and 0.49 ng/mL [1.7, 1.1 and 1.1 pmol/mL] on days 3, 7 and 28, respectively. In infants fed a milk formula containing 68 ng/mL phylloquinone, the plasma concentration rose steadily, with mean values of 1.4, 3.1 and 4.4 ng/mL [3.2, 6.8, and 9.9 pmol/mL] on days 3, 7 and 28, respectively. In another group of infants, Pietersma-de Bruyn et al. (1990) found that phylloquinone was undetectable in cord blood and in venous blood taken at 30 min but became measurable in venous blood after 12 h in 30% of infants (range, 0.04–0.40 ng/mL) and after 24 h in 60% of infants (range, 0.04–0.63 ng/mL).

A more detailed longitudinal comparison of plasma concentrations in breast-fed and formula-fed infants at 6, 12 and 26 weeks was made by Greer et al. (1991). This study is of special interest because the intakes of phylloquinone were also estimated at each time by measuring the vitamin K content of the milk and the volume of milk ingested (by weighing the infant). Such an assessment of the intake of phylloquinone depends on both the analytical accuracy of the measurements in breast milk and validation of the milk collection and sampling technique; both have proved problematical. The study of Greer et al. (1991) seems to have met the requisite criteria, and, although the concentrations were at the lower end of published values, they were in the same range as those in a carefully designed longitudinal study of the phylloquinone content of breast milk over the first five weeks of lactation (1–2 ng/mL) (von Kries et al., 1987b). The results, summarized in Table 8, illustrate the extreme differences in intakes between breast-fed and formula-fed infants, which are also reflected in the plasma concentrations. The plasma concentrations in the formula-fed infants agree with those found by Pietersmade Bruyn et al. (1990) after 28 days (4.5 ng/mL), and suggest that they plateau at around one month. The concentrations in entirely breast-fed infants aged one month and beyond tend, as in this study, to be at the lower end of the normal range in adults (∼0.15–1.0 ng/mL; mean, ∼0.5 ng/mL), even when the infants have received prophylaxis during the first week of life (Cornelissen et al., 1992; Schubiger et al., 1993, 1997). In contrast, the plasma concentrations in formula-fed infants are about 10-fold higher than the average values in adults (Pietersma-de Bruyn et al., 1990; Greer et al., 1991).

Table 8. Dietary intakes and plasma concentrations of phylloquinone in breast-fed and formula-fed infants aged 0–6 months in the USA.

Table 8

Dietary intakes and plasma concentrations of phylloquinone in breast-fed and formula-fed infants aged 0–6 months in the USA.

(g) Hepatic catabolism

The liver plays an exclusive role in the metabolic transformations leading to the elimination of vitamin K from the body. After intravenous doses of 45 µg to 1 mg [3H]phylloquinone, about 20% of the radiolabel was excreted in the urine within three days, and 35–50% was excreted as metabolites in the faeces via the bile (Shearer et al., 1974). Rapid depletion of hepatic reserves of phylloquinone was also seen in surgical patients placed on a low-phylloquinone diet (Usui et al., 1990). These results suggest that the body stores of vitamin K are replenished constantly.

The route of hepatic catabolism leading to urinary excretion of vitamin K proceeds by oxidative degradation of the phytyl side-chain, probably involving the same enzymes used for ω-methyl and β-oxidation of fatty acids, steroids and prostaglandins. Two major metabolites or aglycones have been identified, which are carboxylic acids with five- and seven-carbon atom side-chains and are excreted in the urine as glucuronide conjugates (McBurney et al., 1980). The biliary metabolites have not been clearly identified but are initially excreted as water-soluble conjugates and become lipid-soluble during their passage through the gut, probably through deconjugation by the gut flora. There is no evidence that the body stores of vitamin K are conserved by enterohepatic circulation. Vitamin K itself is too lipophilic to be excreted in the bile, and the side-chain-shortened carboxylic acid metabolites are not biologically active.

(h) Vitamin K-epoxide cycle

In all tissues and cells found to carry out vitamin K-dependent carboxylation, the reaction has been shown to be intimately linked to a metabolic sequence known as the vitamin K-epoxide cycle. This cycle and the associated enzyme activities are shown in Figure 1. Its function seems to be to serve as a salvage pathway to conserve tissue reserves of vitamin K. In the course of γ-glutamyl carboxylation, vitamin K quinol is transformed into vitamin K epoxide, and the epoxide product is recycled in two steps; firstly by vitamin K epoxide reductase activity to produce vitamin K quinone and secondly by quinone reductase activity to produce the co-enzyme vitamin K quinol. Both these activities are thiol-dependent and are probably effected by the same enzyme (Suttie, 1987).

An important property of the dithiol-dependent epoxide and quinone reductase is their sensitivity to certain antagonists, especially those based on 4-hydroxycoumarin (e.g. warfarin) or indandione structures, which have long been used as oral anticoagulants. It is now clear that their anticoagulant action is based on their ability to inhibit epoxide reductase activity and block the recycling of the vitamin. The dithioldependent quinone reductase is also sensitive to warfarin, but the activity of a second quinone reductase catalysed by an NAD(P)H-dependent enzyme is less sensitive to warfarin inhibition and provides an alternative pathway for the reduction of vitamin K quinone to quinol in the presence of warfarin and other oral anticoagulant drugs (Shearer, 1992).

(i) Menadione and related water-soluble derivatives

No studies appear to have been conducted on the absorption, distribution, metabolism or excretion of menadione and related compounds in humans. Water-soluble salts of menadione (vitamin K3) were introduced for vitamin K prophylaxis in newborns in the early 1940s and, until their use was almost entirely superseded by phylloquinone in the early 1960s, there were no suitable techniques for measuring menadione, its salts or their metabolites other than by radioisotopic techniques.

4.1.2. Experimental systems
(a) Absorption

The route and mechanism of absorption of menadione is different from that of natural K vitamins such as phylloquinone. Jaques et al. (1954) fed [14C]menadione to rats and measured the radiolabel in faeces, bile, lymph and urine. They deduced that all the absorbed menadione was transported exclusively via the portal vein to the liver, unlike phylloquinone which is transported by the lymphatic pathway. Also unlike phylloquinone, menadione participated in rapid entero-hepatic circulation after excretion in the bile. Mezick et al. (1968) suggested that, while the portal route is important in rats, menadione could also be transported via the lymphatic system. Direct evidence for some lymphatic transport was found by experiments in dogs, showing that about 10% of the absorbed menadione was recovered in thoracic duct lymph. In studies with bile exclusion, the absorption of menadione in rats was found not to be dependent on bile, as would be expected if menadione is absorbed predominantly via the portal vein.

(b) Tissue distribution, metabolism and excretion

Early experiments with [14C]menadione in mice showed rapid clearance from the intramuscular injection site of doses of 0.1 and 1.0 mg (about 4–40 mg/kg bw) within the first hour and excretion in the urine. Radiolabel was initially detectable in blood, but the concentrations later declined. No significant accumulation was seen in tissues. Small amounts of activity were sometimes detected in liver, lung and kidney, but no significant amounts were found in skin, bone or muscle (Solvonuk et al., 1952). A comparison of the tissue distribution of [14C]menadione and [14C]phylloquinone in rats after intravenous administration of a pharmacological dose (5 mg/kg bw) showed a much higher (24-fold) concentration of radiolabel in the livers of animals given phylloquinone than in those given menadione, and a fivefold greater accumulation of phylloquinone was found in the spleen. As in the studies of Solvonuk et al. (1952), no organ-specific accumulation of radiolabel was found in rats given labelled menadione, the highest proportions of radioactivity being found in urine and faeces (Taylor et al., 1957). The rapid, extensive excretion of [14C]menadione in the urine was confirmed by Losito et al. (1968) who found that rats excreted about 70% of an intravenous dose in the urine within 24 h compared with only about 10% of a dose of phylloquinone. They also showed that the urinary excretion of menadione (again unlike phylloquinone) was not dependent on an intact liver, as hepatectomized rats excreted the same amount of the dose (70%) as normal rats.

Rats given intracardial injections of a more physiological total dose (10 µg [30 µg/kg bw]) of high-activity 6,7-[3H]menadione showed a pattern of excretion and tissue distribution similar to that of pharmacological doses, with recovery of 78–83% of the label in the urine after 18 h (Taggart & Matschiner, 1969). A similar pattern was seen in rats given an intraperitoneal injection of about 2 µg of the water-soluble salt menadiol diphosphate; 17 h later, some 43% of the radiolabel had been excreted in urine and about 4% in faeces. The compound was not concentrated in any tissue but was distributed throughout all body organs, and the distribution was the same in vitamin K-replete and -deficient animals. This water-soluble compound underwent rapid conversion to lipid-soluble forms, and the compound and its metabolites were found generally to be associated with the membranous fractions of cells (Thierry & Suttie, 1969).

Three major metabolites of menadione have been isolated from urine. After oral administration of menadione to rabbits, Richert (1951) isolated the sulfated compound 2-methyl-4-hydroxy-1-naphthyl sulfate and noted increased excretion of glucuronic acid. Hoskin et al. (1954) resolved three metabolites from rat urine, of which the major product was tentatively identified as 2-methyl-1,4-dihydroxynaphthalene-1,4-diglucuronide and another as the monosulfate conjugate found by Richert (1951). A third, minor metabolite appears to be a phosphate conjugate (Hart, 1958).

Losito et al. (1967) showed in an isolated perfused rat liver system that menadione glucuronide or sulfate conjugates are excreted but that the glucuronide is confined to bile and the sulfate to the perfusing blood. In rats in vivo, Losito et al. (1968) separated three major urinary metabolites, two of which were identified as the same glucuronide and sulfate conjugates as those found in their liver perfusion system (Losito et al. 1967). The chromatographic pattern in hepatectomized rats was different, but the major peak was shown to be a glucuronide conjugate, showing that animals have the capacity for extrahepatic conjugation of menadione with glucuronic acid (Losito et al., 1968).

(c) Conversion of menadione to menaquinone-4

The vitamin K activity of menadione and its water-soluble salts depends on its specific metabolic conversion to menaquinone-4 (Suttie, 1985, 1991). The early evidence that both menadione and phylloquinone could be converted in birds and rats has been reviewed (Martius, 1967). The enzymic alkylation of menadione to menaquinone-4 was subsequently confirmed by more sophisticated techniques both in vivo in rats (Taggart & Matschiner, 1969) and in vitro in chick liver homogenates (Dialameh et al., 1970). The greatest alkylating activity was found in the microsomal fraction and was six to seven times higher in chick liver microsomes than in rat liver microsomes (Dialameh et al., 1970).

4.2. Toxic effects

4.2.1. Humans
(a) Phylloquinone

Reports of acute toxicity associated with pharmaceutical preparations of vitamin K as phylloquinone are rare and are often attributed to the vehicle of solubilization or other component of the preparation rather than to vitamin K itself. Adverse events associated with two products (Konakion and Konakion MM, currently representing about 50% of the market share worldwide) were monitored in a post-marketing surveillance programme, and the results were analysed and reviewed by Pereira and Williams (1998). During the period 1974 to July 1995, an estimated 635 million adults and 728 million children were prescribed Konakion or Konakion MM, and only 404 adverse events in 286 subjects were reported. Of these, the majority (96%) were associated with the older, Cremophor EL-based Konakion, which accounted for 95% of sales during this period. ‘Skin, hair and nail disorders’ were the most common adverse effects, accounting for about 25% of those reported. Rare cutaneous reactions to another vitamin K preparation, AquaMephyton, have been reported and are suspected to be immunologically mediated (Sanders & Winkelmann, 1988). This preparation contains a polyoxyethylated fatty acid derivative as the emulsifying agent (Rich & Drage, 1982).

The most serious reaction to vitamin K is anaphylactoid reactions after parenteral administration, but evidence that this effect is due to the polyethoxylated castor oil emulsifier (non-ionic detergent) Cremophor EL (polyethyleneglycolglycerol riconoleate) rather than vitamin K is twofold. Firstly, during the last 12 months of post-marketing surveillance (1994–95), 14 serious adverse events were reported from an estimated 21 million individuals receiving the Cremophor EL-based Konakion but none from the 13 million who received Konakion MM (Pereira & Williams, 1998). Secondly, anaphylactoid reactions in humans have been reported with other drugs solubilized with Cremophor EL, and there is experimental evidence in dogs that Cremophor EL and its components cause histamine release and hypotensive reactions (Lorenz et al., 1982). The mixed-micellar Konakion MM preparation in which the vitamin K is solubilized by the naturally occurring components glycocholic acid and phosphatidylcholine appears to have far fewer anaphylactoid properties, only one probable anaphylactoid reaction having been reported in an estimated 66 million adults and 1–2 million infants and children who received this preparation (Pereira & Williams, 1998). Severe complications resulting in cardiopulmonary arrest were reported after intravenous injection of AquaMephyton (Rich & Drage, 1982).

(b) Menadione

The potential toxicity of preparations of menadione and its water-soluble derivatives to newborn infants is well established and has been reviewed (Vest, 1966). The toxic reactions commonly include haemolytic symptoms evidenced by increased reticulocyte counts and Heinz body formation. In severe cases, overt haemolytic anaemia with haemoglobinuria may occur. The increased erythrocyte breakdown may lead to hyperbilirubinaemia and kernicterus. These effects are clearly dose-dependent, as premature infants given 30 mg of menadiol sodium phosphate had higher serum bilirubin concentrations, more Heinz bodies, lower haemoglobin concentrations and lower erythrocyte counts than those given 1 mg. The toxic reactions are more pronounced and may lead to severe haemolysis in premature infants and in infants with a congenital defect of glucose 6-phosphate dehydrogenase.

An explanation for the haemolytic toxicity of menadione is provided by studies showing the high reactivity of the 3-position of menadione with sulfhydryl compounds. Canady and Roe (1956) showed that when menadione is added to blood, it combines directly with blood proteins, probably by forming a thio ether at the 3-position. A later study showed that menadione reacts with both the haem groups and the β-93 thiol groups of haemoglobin and that it oxidizes the haem groups of oxyhaemoglobin, resulting in the formation of methaemoglobin (Winterbourn et al., 1979).

With elucidation of the toxic properties of menadione in newborn infants and, in the 1960s, the industrial synthesis of natural K vitamins, use of menadione for vitamin K prophylaxis in the newborn was discontinued in most countries (Vest, 1966).

4.2.2. Experimental systems
(a) Phylloquinone

Israels et al. (1983) suggested that vitamin K compounds may have a regulatory function in the metabolism of benzo[a]pyrene and possibly other compounds that are metabolized through the mixed-function oxidase system. This suggestion stemmed from their studies with menadione, which was shown to inhibit the conversion of benzo[a]pyrene to its more polar metabolites in rat liver microsomes in vitro. The inhibition showed a plateau (25% of control) at a concentration of 100 µmol/L [17 µg/mL]. With phylloquinone, no inhibition to polar metabolites was evident at concentrations up to 50 µmol/L [8.6 µg/mL], but at concentrations of 50–200 µmol/L [34 µg/mL] the inhibition increased rapidly, and at 500 µmol/L [86 µg/mL] the degree of inhibition was similar to that produced by menadione. The authors concluded that menadione acted as an electron acceptor. The weaker effect of phylloquinone at lower concentrations is perhaps due to its much greater lipophilicity and reduced penetration and solubility in microsomal membranes as compared with menadione; this explanation would also be consistent with the absence of a difference in solubility at higher concentrations of phylloquinone. In a later paper, Israels et al. (1985) found that microsomal metabolism of benzo[a]pyrene to polar metabolites in vitro was actually increased when the concentration of phylloquinone was reduced to 25 µmol/L [11.3 µg/mL] but, as in their earlier paper, was decreased at a concentration of 200 µmol/L [90 µg/mL].

In studies of the effects of menadione and phylloquinone on tumorigenesis in mice in vivo, the rate of tumour appearance and the death rate of mice given an intraperitoneal injection of benzo[a]pyrene were slowed by menadione but increased by phylloquinone. In parallel studies, tumorigenesis was inhibited in mice treated with the vitamin K antagonist warfarin and in mice made vitamin K-deficient by dietary deprivation. In these experiments, the compounds were given either before or both before and after benzo[a]pyrene (Israels et al., 1983).

(b) Menadione

Menadione also causes haemolytic anaemia in animals. The results of studies conducted in the 1940s were confirmed by Munday et al. (1991), who gave menadione (in 2% Tween 80) to Sprague-Dawley rats at a single dose of 750 µmol/kg bw per day [equivalent to about 100 mg/kg bw per day] for six consecutive days. This dose resulted in significant increases in splenic weight and decreased blood packed cell volume and haemoglobin concentration. Heinz bodies were observed in stained erythrocytes. There was no evidence that menadione caused haemaglobinaemia, suggesting that the haemolysis is not intravascular but is due to the destruction of damaged erythrocytes by cells of the reticuloendothelial system. Haemolysis was the only toxic change identified in rats dosed with menadione.

Melgar et al. (1991) examined the toxicity of menadione by giving Sprague-Dawley rats gradually increasing oral doses of menadione for six weeks, starting at 5 mg/kg bw per day and increasing to 20 mg/kg bw per day in the third week and 40 mg/kg bw per day in the fifth week of treatment. This dose regime was generally well tolerated with no relevant haematological changes, although there was a significant increase in spleen weight.

Many studies have been reported of the cytotoxicity of menadione in isolated and cultured cells of several types, including isolated rat hepatocytes (Mirabelli et al., 1988; Shertzer et al., 1992; Toxopeus et al., 1993), rat renal epithelial cells (Brown et al., 1991), bovine heart microvascular endothelial cells (Kossenjans et al., 1996), Chinese hamster V79 cells (Ochi, 1996) and human hepatoma and leukaemia cell lines (Chiou et al., 1998). The cytotoxicity of menadione has also been studied in isolated rat platelets (Chung et al., 1997).

A characteristic finding in isolated rat hepatocytes treated with menadione is the appearance of numerous protrusions in the plasma membrane, known as blebs. Menadione produced a dose- and time-dependent increase in the frequency of cytoskeletal abnormalities; protein thiol oxidation seems to be intimately related to the appearance of surface blebs (Mirabelli et al., 1988).

4.3. Reproductive and prenatal effects

4.3.1. Humans

No formal investigations of the safety of vitamin K in pregnancy have been found, although it has been proposed that vitamin K deficiency causes congenital malformations (Menger et al., 1997). In a study of the efficacy of vitamin K for the prevention of the vitamin K deficiency induced by antiepileptic drugs, 16 women receiving antiepileptic drugs known to induce hepatic microsomal enzymes were treated orally with phylloquinone (Konakion) at 10 mg/day from the 36th week of pregnancy until delivery (mean, 29 days; range, 10–46). A control group of 20 epileptic women on similar antiepileptic drugs did not receive supplemental vitamin K. No adverse effects were observed in the infants of women given vitamin K supplementation. The median maternal plasma concentration of phylloquinone was raised 60-fold and the cord blood concentration was raised 15-fold, for a median maternal:cord blood ratio of 44 (Cornelissen et al., 1993b).

4.3.2. Experimental systems

The offspring of mice treated with phylloquinone by injection had cleft lip and exencephaly (Schardein, 1993). Six pregnant Sprague-Dawley rats were dosed with 10 mg/kg bw phylloquinone (Konakion) daily on days 9–20 of gestation, and the fetuses were delivered on day 21 and examined for external malformations and the presence of haemorrhages only. No adverse effects were noted when compared with a group of five untreated controls (Howe & Webster, 1990). [The Working Group noted the small numbers of animals and the restricted fetal examination.]

Oral administration of menadione to groups of 10 pregnant Wistar rats throughout gestation at a dose of 0.15, 15 or 150 mg/day [approximately 0.6, 60 or 600 mg/kg bw per day] had no adverse effect on maternal body-weight gain, pregnancy rate or litter size, but the fetuses showed slightly retarded growth and delayed ossification at the high dose. No abnormalities were observed (Kosuge, 1973).

4.4. Genetic and related effects

4.4.1. Humans

Cornelissen et al. (1991) observed no difference in sister chromatid exchange or chromosomal aberration frequency in peripheral blood lymphocytes from six neonates given intramuscular phylloquinone prophylaxis and in those from six control neonates. The blood was taken 24 h after an intramuscular dose of 1 mg, at which time the plasma concentrations of phylloquinone ranged from 115 to 1150 ng/mL (mean, 536 ng/mL), compared with about 0.15 ng/mL in the control neonates.

Pizer et al. (1995) used the glycophorin A mutation assay to assess the risk for somatic mutations of NO and NN variant red cells of 64 infants aged 10 days to six months heterozygous for the MN blood group, who had received either oral, intramuscular or intravenous phylloquinone prophylaxis at birth. All three groups showed a lower variant frequency than a reference group of children aged 1–15 years. For ethical reasons, there was no control group of infants who had not received vitamin K prophylaxis, and the conclusion was therefore limited to a lack of association between the route of vitamin K administration and somatic mutation.

4.4.2. Experimental systems

Limited data are available on the genetic and related effects of phylloquinone and menaquinones (Table 9). Phylloquinone did not induce mutation in Salmonella typhimurium. It enhanced the frequency of sister chromatid exchange in cultured human maternal lymphocytes at concentrations that are relevant in vivo, and a similar increase in sister chromatid exchange frequency was observed in cultured lymphocytes from human placental blood. In fetal sheep that received a catheter in the femoral vein 10–15 days before term, phylloquinone significantly increased the frequency of sister chromatid exchange in peripheral blood lymphocytes sampled 24 h later.

Table 9. Genetic and related effects of phylloquinone and menadione.

Table 9

Genetic and related effects of phylloquinone and menadione.

Menaquinone-4 but not phylloquinone inhibited osteoclastic bone resorption by inducing osteoclast apoptosis (Kameda et al., 1996). Menaquinone-4 and its derivatives also induced apoptosis in various human leukaemic cell lines (Yaguchi et al., 1997).

In preincubation protocols with Ames Salmonella tester strains, menadione did not induce reverse mutation in strains TA100, TA102, TA1535, TA1537, TA1538 or TA2638 in the presence or absence of an exogenous metabolic activation system. It was mutagenic in TA98 with metabolic activation and in TA2637 with or without activation. Menadione also induced mutation in strain TA104, but only with metabolic activation by purified NADPH–cytochrome P450 reductase; in another study it was mutagenic in this strain without activation. Menadione did not induce reverse mutation in Escherichia coli WP2/pKM101 or WP2uvrA/pkM101 in the absence of metabolic activation.

In tests with derivatives of E. coli WP2s (uvrA trpE) that are defective in 7,8-dihydro-8-oxoguanine DNA glycosylase activity (mutM) or MutY glycosylase activity on an A:7,8-dihydro-8-oxoguanine mispair (mutY) or give an adaptive response to oxidative stress by superoxide (soxRS), to compare the mutability of various reactive oxygen-generating compounds, menadione was not mutagenic; however, it was mutagenic in two strains of E. coli WP2 that contain deficiencies in the oxyR function. Menadione induced forward mutation to l-arabinose resistance (AraR) in E. coli K-12 strains with diminished concentrations of superoxide dismutase and induced a SOS response in PQ37.

This agent induced concentration-dependent single-strand and double-strand DNA breaks in a human breast cancer MCF-7 cell line, in cultured rat hepatocytes, in human fibroblasts, in human chronic myeloid leukaemic K562 cells and in a single-cell gel electrophoresis assay to measure DNA strand breaks in human lymphocytes at doses as low as 1 µmol/L. At concentrations of 15–100 µmol/L, menadione induced extensive DNA fragmentation in human chronic myeloid leukaemic K562 cells which could be measured in alkaline elution assays. At these doses, no oxidative stress appeared to occur in these cells.

Cantoni et al. (1991) reported that hydrogen peroxide produced during the metabolism of menadione does not contribute to the cytotoxic action of the quinone. In isolated rat hepatocytes, menadione induced DNA fragmentation consistent with apoptosis. These effects occurred in the absence of 8-oxo-2′-deoxyguanosine production, and the authors concluded that oxidative modification of DNA bases was unlikely to be involved (Fischer-Nielsen et al., 1995). Menadione induced protein-linked DNA breaks in the presence of purified human DNA topoisomerase II but not DNA topoisomerase I (Frydman et al., 1997), and it seems likely that DNA topoisomerase II poisoning is involved in DNA breakage by menadione at the lower concentrations, at which oxygen stress does not occur.

Menadione induced morphological transformation of BALB/c 3T3 cells, but only when tested in the presence of the tumour promotor 12-O-tetradecanoylphorbol 13-acetate.

Andrew et al. (1999) found that menadione enhanced the spontaneous mutation frequency and induced a novel mutation spectrum of lacI genes recovered from a rat embryonic fibroblast line transfected with a λ-phage shuttle vector, in both the traditional plaque assay and a positive selection assay.

4.5. Mechanistic considerations

(a) Phylloquinone

On the basis of studies of microsomal metabolism in vitro and studies in rats and mice in vivo, Israels et al. (1983, 1985) suggested that vitamin K may be mutagenic by affecting the mixed-function oxidase system which metabolizes benzo[a]pyrene. Phylloquinone at a high concentration (200 µmol/L) inhibited the conversion of benzo[a]pyrene to its more polar metabolites, a property it shares with menadione. Paradoxically, at a lower concentration of phylloquinone (25 µmol/L), but not with menadione, the metabolism of benzo[a]pyrene was increased. In this system, therefore, whereas menadione consistently acts as a potential inhibitor of carcinogenesis, phylloquinone could either potentiate or inhibit it, depending on the concentration. The overall weaker inhibitory effect of phylloquinone could be due to the low solubility of this lipophilic compound, but it is difficult to explain the mechanism of the enhanced metabolism of benzo[a]pyrene at lower concentrations of phylloquinone.

In studies in vivo, Israels and co-workers found that menadione and vitamin K deficiency (nutritional or induced by the vitamin K antagonist, warfarin) both inhibited the rates of benzo[a]pyrene-induced tumour appearance and death, whereas phylloquinone increased the rate of carcinogenesis. They concluded that vitamin K deficiency confers a protective effect against benzo[a]pyrene-induced tumour formation. They subsequently tendered the hypothesis that the low vitamin K status of normal newborns confers a biological advantage by reducing the risk of mutagenic events during a period of rapid cell proliferation (Israels et al., 1987; Saxena et al., 1997).

Vervoort et al. (1997) reported that metabolic cycling of vitamin K compounds via the vitamin K cycle (Figure 1) confers potent antioxidant activity against lipid peroxidation. They concluded that the antioxidant effect is probably due to radical chain-breaking by vitamin K quinol and that dietary intake of vitamin K may strengthen cellular defences against oxidative stress.

(b) Menadione

In many of the studies of the cytotoxicity of menadione in cultured cells and blood platelets, menadione was used as a model compound for induction of cellular damage either by arylating protein-bound and soluble thiols or by inducing oxidative stress. The relative importance of these two mechanisms is difficult to determine. The toxicity may result directly from binding of menadione to a critical protein thiol (such as a membrane cation transporter) or indirectly from binding to and decreasing concentrations of reduced glutathione, thereby predisposing the cell to oxidative stress. An alternative mechanism whereby menadione may produce oxidative stress is by redox cycling, which ultimately results in the production of reactive oxygen species. Oxidative stress results when the production of reactive oxygen species exceeds the antioxidant defence mechanisms, which in turn may result in cellular injury and death through a variety of mechanisms. In human cancer cells, menadione-induced cell degeneration was considered to result mainly from lipid peroxidative damage rather than from other mechanisms such as a depleted glutathione content (Chiou et al., 1998).

It has been proposed that menadione causes mutations by generating active oxygen species from semiquinone radicals (e.g. Chesis et al., 1984; Smith et al., 1987; Hakura et al., 1994; Morgan et al., 1998). Semiquinones can generate superoxide anion, which itself produces other active species, such as hydrogen peroxide and hydroxyl radical, through enzyme- and metal-catalysed reactions (Chesis et al., 1984).

It now seems likely that menadione has an additional mode of action as a mutagen, by acting as a poison of DNA topoisomerase II enzymes. This could well be responsible for the DNA breakage, chromosomal aberrations and apoptosis observed in mammalian cells under conditions that did not lead to oxidative stress (e.g. Sawada et al., 1987; Fischer-Nielsen et al., 1995; Morgan, 1995). Cells in culture can, however, convert menadione to menaquinone-4, and there is already evidence that this plays a role in apoptosis.

5. Summary of Data Reported and Evaluation

5.1. Exposure data

The term ‘vitamin K’ refers to a group of 2-methyl-1,4-naphthoquinone derivatives which can fulfil an essential co-factor function in humans in the biosynthesis of a number of calcium-binding proteins, some of which are essential for haemostasis. In nature, vitamin K occurs as phylloquinone in plants and as menaquinones produced by bacteria. The major dietary sources of vitamin K are green leafy vegetables and certain vegetable oils. Clinically, vitamin K is used primarily to prevent or cure deficiency-related bleeding in newborns and patients with malabsorption syndromes and to reverse the anticoagulative effects of vitamin K antagonists.

5.2. Human carcinogenicity data

An association between childhood leukaemia and vitamin K prophylaxis given by the intramuscular route was found in two reports but was not confirmed in a number of studies in various countries. A major limitation of most of the studies is that the fact of intramuscular administration of vitamin K was difficult to establish retrospectively for a substantial proportion of subjects, although the results of the analyses based on individual records and on imputed hospital policies for vitamin K administration are similar. In the studies in which a suggestion of an association was observed, selection bias may have accounted for the result. The possibility cannot be entirely excluded of a small increase in the risk for acute lymphoblastic leukaemia occurring at ages around those of the peak incidence in childhood in children given intramuscular administration of vitamin K.

The few studies that investigated oral administration of vitamin K found no increase in the relative risk for leukaemia.

5.3. Animal carcinogenicity data

No adequate study on the carcinogenicity of vitamin K substances was available to the Working Group.

5.4. Other relevant data

Phylloquinone and menaquinones are absorbed from food into the lymphatic system and carried by triglyceride-rich lipoproteins in the blood. Menaquinones synthesized by the gut microflora may also be absorbed. Phylloquinone is rapidly cleared from the circulation by the liver, metabolized to metabolites with shortened side-chains and excreted in the bile and urine. In animals, menadione is absorbed predominantly by the portal route, does not accumulate in specific organs and is extensively excreted unchanged in the urine. A fraction of menadione is converted in tissues to menaquinone-4.

Phylloquinone rarely has toxic effects, and the few serious immunological complications observed have been attributed to the vehicle of solubilization. Menadione may cause haemolytic anaemia and induce cellular damage by arylating protein-bound and soluble thiols or by inducing oxidative stress.

No adverse effects have been reported in mothers or infants after administration of vitamin K during pregnancy, whereas vitamin K deficiency is teratogenic. The safety of vitamin K in pregnancy has not been adequately studied experimentally.

Neither phylloquinone nor menaquinones have been adequately studied for mutagenicity. Menadione acts as a bacterial mutagen in several specific strains of Salmonella typhimurium and Escherichia coli. In mammalian cells, menadione leads to DNA breakage, and there are isolated reports of chromosomal aberrations and sister chromatid exchange.

5.5. Evaluation

There is inadequate evidence in humans for the carcinogenicity of vitamin K substances.

There is inadequate evidence in experimental animals for the carcinogenicity of vitamin K substances.

Overall evaluation

Vitamin K substances are not classifiable as to their carcinogenicity to humans (Group 3).

6. References

  • American Hospital Formulary Service (1997) AHFS Drug Information® 97, Bethesda, MD, American Society of Health-System Pharmacists, pp. 2834–2836.
  • Andrew S.E., Hsiao L., Milhausen K., Jirik F.R. Comparison of selectable and plaque assay systems to detect menadione- and UV-induced lacI mutations in mammalian cells. Mutat. Res. 1999;427:89–97. [PubMed: 10393263]
  • van den Anker J.N., Sinaasappel M. Bleeding as presenting symptom of cholestasis. J. Pernatol. 1993;13:322–324. [PubMed: 8410391]
  • Ansell P., Bull D., Roman E. Childhood leukaemia and intramuscular vitamin K: Findings from a case–control study. Br. med. J. 1996;313:204–205. [PMC free article: PMC2351604] [PubMed: 8696197]
  • AOAC International (1996) AOAC Official Method 992.27. trans-Vitamin K1 (phylloquinone) in ready-to-feed milk-based infant formula. In: Official Methods of Analysis of AOAC International, 16th Ed., 4th rev., Gaithersburg, MD [CD-ROM edition]
  • Blanco M., Urios A., Martínez A. New Escherichia coli WP2 tester strains highly sensitive to reversion by oxidative mutagens. Mutat. Res. 1998;413:95–101. [PubMed: 9639684]
  • Booth S.L, Suttie J.W. Dietary intake and adequacy of vitamin K1. J. Nutr. 1998;128:785–788. [PubMed: 9566982]
  • Booth S.L., Sadowski J.A., Pennington J.A.T. Phylloquinone (vitamin K1) content in the US Food and Drug Administration’s Total Diet Study. J. agric. Food Chem. 1995;43:1574–1579.
  • Booth S.L., Pennington J.A.T., Sadowski J.A. Food sources and dietary intakes of vitamin K-1 (phylloquinone) in the American diet: Data from the FDA Total Diet Study. J. Am. diet. Assoc. 1996;96:149–154. [PubMed: 8557941]
  • Breen G.A., St Peter W.L. Hypoprothombinemia associated with cefmetazole. Ann. Pharmacother. 1997;31:180–184. [PubMed: 9034420]
  • British Medical Association/Royal Pharmaceutical Society of Great Britain (1998) British National Formulary, No. 36, London, pp. 422–423.
  • British Pharmacopoeial Commission (1993) British Pharmacopoeia 1993, London, Her Majesty’s Stationery Office, Vols I & II, pp. 409–411, 516–517, 999–1000, 1059–1060, S79–S80.
  • Brown P.C., Dulik D.M., Jones T.W. The toxicity of menadione (2-methyl-1,4-naphthoquinone) and two thioether conjugates studied with isolated renal epithelial cells. Arch. Biochem. Biophys. 1991;285:187–196. [PubMed: 1990978]
  • Budavari, S., ed. (1996) The Merck Index, 12th Ed., Whitehouse Station, NJ, Merck & Co., pp. 994, 1269–1270.
  • Bueno M.P., Villalobos M.C. Reverse phase high pressure liquid chromatographic determination of vitamin K1 in infant formulas. J. Assoc. off. anal. Chem. 1983;66:1063–1066. [PubMed: 6630121]
  • Canadian Pharmaceutical Association (1997) CPS Compendium of Pharmaceuticals and Specialties, 32nd Ed., Ottawa, Ontario, p. 1730.
  • Canady W.J., Roe J.H. Studies on the reaction of menadione with blood and denatured proteins. J. biol. Chem. 1956;220:571–582. [PubMed: 13331916]
  • Canfield, L.M., Martin, G.S. & Sugimoto, K. (1988) Vitamin K in human milk. In: Suttie, J.W., ed., Current Advances in Vitamin K Research, New York, Elsevier, pp. 499–504.
  • Cantoni O., Fiorani M., Cattabeni F., Bellomo G. DNA breakage caused by hydrogen peroxide produced during the metabolism of 2-methyl-1,4-naphthoquinone (menadione) does not contribute to the cytotoxic action of the quinone. Biochem. Pharmacol. 1991;42:S220–S222. [PubMed: 1662950]
  • Carstensen J. Intramuscular vitamin K and childhood cancer (Letter to the Editor). Br. med. J. 1992;305:709–710. [PMC free article: PMC1882958] [PubMed: 1393124]
  • Chesis P.L., Levin D.E., Smith M.T., Ernster L., Ames B.N. Mutagenicity of quinones: Pathways of metabolic activation and detoxification. Proc. natl Acad. Sci. USA. 1984;81:1696–1700. [PMC free article: PMC344985] [PubMed: 6584903]
  • Chiou T.-J., Chou Y.-T., Tzeng W.-F. Menadione-induced cell degeneration is related to lipid peroxidation in human cancer cells. Proc. natl Sci. Counc., ROC, Part B: Life Sci. 1998;22:13–21. [PubMed: 9536516]
  • Choo K.E., Tan K.K., Chuah S.P., Ariffin W.A., Gururaj A. Haemorrhagic disease in newborn and older infants: A study in hospitalized children in Kelantan, Malaysia. Ann. trop. Paediatr. 1994;14:231–237. [PubMed: 7825997]
  • Chung J.-H., Seo D.-C., Chung S.-H., Lee J.-Y., Seung S.-A. Metabolism and cytotoxicity of menadione and its metabolite in rat platelets. Toxicol. appl. Pharmacol. 1997;142:378–385. [PubMed: 9070361]
  • CIS Information Services (1998) Worldwide Bulk Drug Users Directory 1997/98 Edition, Dallas, TX [CD-ROM]
  • Clark F.I., James E.J. Twenty-seven years of experience with oral vitamin K1 therapy in neonates. J. Pediatr. 1995;127:301–304. [PubMed: 7636660]
  • Conly J.M., Stein K. Quantitative and qualitative measurements of K vitamins in human intestinal contents. Am. J. Gastroenterol. 1992;87:311–316. [PubMed: 1539565]
  • Cook A., Stovicek R., D’Odorico A., Tkac A., Bilton R.F. Menaquinone mediated free radical generation: A possible mutagenic mechanism. Biochem. Soc. Trans. 1991;19:426S. [PubMed: 1665440]
  • Cornelissen M., Smeets D., Merkx G., De Abreu R., Kollée L., Monnens L. Analysis of chromosome aberrations and sister chromatid exchanges in peripheral blood lymphocytes of newborns after vitamin K prophylaxis at birth. Pediatr. Res. 1991;30:550–553. [PubMed: 1805152]
  • Cornelissen E.A.M., Kollée L.A.A., De Abreu R.A., van Baal J.M., Motohara K., Verbruggen B., Monnens L.A.H. Effects of oral and intramuscular vitamin K prophylaxis on vitamin K1, PIVKA-II, and clotting factors in breast fed infants. Arch. Dis. Child. 1992;67:1250–1254. [PMC free article: PMC1793939] [PubMed: 1444522]
  • Cornelissen E.A.M., van Lieburg A.F., Motohara K., van Oostrom C.G. Vitamin K status in cystic fibrosis. Acta paediatr. 1992;81:658–661. [PubMed: 1421902]
  • Cornelissen M., Steegers-Theunissen R., Kollée L., Eskes T., Vogels-Mentink G., Motohara K., De Abreu R., Monnens L. Increased incidence of neonatal vitamin K deficience resulting from maternal anticonvulsant therapy. Am. J. Obstet. Gynecol. 1993a;168:923–928. [PubMed: 8456903]
  • Cornelissen M., Steegers-Theunissen R., Kollée L., Eskes T., Motohara K., Monnens L. Supplementation of vitamin K in pregnant women receiving anticonvulsant therapy prevents neonatal vitamin K deficiency. Am. J. Obstet. Gynecol. 1993b;168:884–888. [PubMed: 8456897]
  • Cornelissen M., von Kries R., Loughnan P., Schubiger G. Prevention of vitamin K deficiency bleeding: Efficacy of different multiple oral dose schedules of vitamin K. Eur. J. Pediatr. 1997;156:126–130. [PubMed: 9039517]
  • Council of Europe (1997) European Pharmacopoeia, 3rd Ed., Strasbourg, pp. 1154–1155, 1332–1333.
  • Crowther M.A., Donovan D., Harrison L., McGinnis J., Ginsberg J. Low-dose oral vitamin K reliably reverses over-anticoagulation due to warfarin. Thromb. Haemostasis. 1998;79:1116–1118. [PubMed: 9657434]
  • Davidson R.T., Foley A.L., Engelke J.A., Suttie J.W. Conversion of dietary phylloquinone to tissue menaquinone-4 in rats is not dependent on gut bacteria. J. Nutr. 1998;128:220–223. [PubMed: 9446847]
  • Denda A., Sai K.M., Tang Q., Tsujiuchi T., Tsutsumi M., Amanuma T., Murata Y., Nakae D., Maruyama H., Kurokawa Y., Konishi Y. Induction of 8-hydroxydeoxyguanosine but not initiation of carcinogenesis by redox enzyme modulations with or without menadione in rat liver. Carcinogenesis. 1991;12:719–726. [PubMed: 1707352]
  • Dialameh G.H., Yekundi K.G., Olson R.E. Enzymatic alkylation of menaquinone-0 to menaquinones by microsomes from chick liver. Biochim. biophys. Acta. 1970;223:332–338. [PubMed: 4323518]
  • Doran O., Austic N.C., Taylor B.J. Vitamin K administration in neonates: Survey of compliance with recommended practices in the Dunedin area. N.Z. J. Med. 1995;108:337–339. [PubMed: 7566761]
  • Draper G., McNinch A. Vitamin K for neonates: The controversy. Br. med. J. 1994;308:867–868. [PMC free article: PMC2539849] [PubMed: 8173359]
  • Draper G.J., Stiller C.A. Intramuscular vitamin K and childhood cancer (Letter to the Editor). Br. med. J. 1992;305:709. [PMC free article: PMC1882918] [PubMed: 1393123]
  • Durie P.R. Vitamin K and the management of patients with cystic fibrosis. Can. med. Assoc. J. 1994;151:933–936. [PMC free article: PMC1337279] [PubMed: 7922929]
  • Editions du Vidal (1998) Dictionnaire Vidal 1998, 74th Ed., Paris, OVP, pp. 1989–1990.
  • Ekelund H. Late haemorrhagic disease in Sweden 1987–89. Acta paediatr. scand. 1991;80:966–968. [PubMed: 1755307]
  • Ekelund H., Finnström O., Gunnarskog J., Källén B., Larsson Y. Administration of vitamin K to newborn infants and childhood cancer. Br. med. J. 1993;307:89–91. [PMC free article: PMC1693492] [PubMed: 8343734]
  • Ferland G., Sadowski J.A. Vitamin K1 (phylloquinone) content of edible oils: Effects of heating and light exposure. J. agric. Food Chem. 1992;40:1869–1873.
  • Fetrow C.W., Overlock T., Leff L. Antagonism of warfarin-induced hypoprothrombinemia with use of low-dose subcutaneous vitamin K1. J. clin. Pharmacol. 1997;37:751–757. [PubMed: 9378848]
  • Fischer-Nielsen A., Corcoran G.B., Poulsen H.E., Kamendulis L.M., Loft S. Menadione-induced DNA fragmentation without 8-oxo-2′-deoxyguanosine formation in isolated rat hepatocytes. Biochem. Pharmacol. 1995;49:1469–1474. [PubMed: 7763290]
  • Food & Drug Administration (1999) Food and drugs. US Code Fed. Regul., Title 21, Part 107.100, Subpart D, pp. 186–187.
  • Fournier B., Sann L., Guillaumont M., Leclercq M. Variations of phylloquinone concentration in human milk at various stages of lactation and in cow’s milk at various seasons. Am. J. clin. Nutr. 1987;45:551–558. [PubMed: 3825982]
  • Frydman B., Marton L.J., Sun J.S., Neder K., Witiak D.T., Liu A.A., Wang H.-M., Mao Y., Wu H.-Y., Sanders M.M., Liu L.F. Induction of DNA topoisomerase II-mediated DNA cleavage by beta-lapachone and related naphthoquinones. Cancer Res. 1997;57:620–627. [PubMed: 9044837]
  • Gennaro, A.R. (1985) Remington’s Pharmaceutical Sciences, 17th Ed., Easton, PA, Mack Publishing Co., pp. 1010–1011.
  • Gennaro, A.R. (1995) Remington: The Science and Practice of Pharmacy, 19th Ed., Easton, PA, Mack Publishing Co., Vol. II, pp. 1114–1115.
  • Gijsbers B.L.M.G., Jie K.-S.G., Vermeer C. Effect of food composition on vitamin K absorption in human volunteers. Br. J. Nutr. 1996;76:223–229. [PubMed: 8813897]
  • Golding J., Paterson M., Kinlen L.J. Factors associated with childhood cancer in a national cohort study. Br. J. Cancer. 1990;62:304–308. [PMC free article: PMC1971807] [PubMed: 2386748]
  • Golding J., Greenwood R., Birmingham K., Mott M. Childhood cancer, intramuscular vitamin K, and pethidine given during labour. Br. med. J. 1992;305:341–346. [PMC free article: PMC1883000] [PubMed: 1392886]
  • Greer F.R., Marshall S., Cherry J., Suttie J.W. Vitamin K status of lactating mothers, human milk, and breast-feeding infants. Pediatrics. 1991;88:751–756. [PubMed: 1896278]
  • Greer F.R., Marshall S.P., Foley A.L., Suttie J.W. Improving the vitamin K status of breastfeeding infants with maternal vitamin K supplements. Pediatrics. 1997;99:88–92. [PubMed: 8989344]
  • Guillaumont M., Sann L., Leclercq M., Dostalova L., Vignal B., Frederich A. Changes in hepatic vitamin K1 levels after prophylactic administration to the newborn. J. pediatr. Gastroenterol. Nutr. 1993;16:10–14. [PubMed: 8433228]
  • Hagstrom J.N., Bovill E.G., Soll R.F., Davidson K.W., Sadowski J.A. The pharmacokinetics and lipoprotein fraction distribution of intramuscular vs. oral vitamin K1 supplementation in women of childbearing age: Effects on hemostasis. Thromb. Haemostasis. 1995;74:1486–1490. [PubMed: 8772225]
  • Hakura A., Mochida H., Tsutsui Y., Yamatsu K. Mutagenicity and cytotoxicity of naphthoquinones for Ames Salmonella tester strains. Chem. Res. Toxicol. 1994;7:559–567. [PubMed: 7981421]
  • Hanawa Y. Vitamin K deficiency in infancy: The Japanese experience. Acta paediatr. jpn. 1992;34:107–116. [PubMed: 1621515]
  • Hansen K.B., Ebbesen F. Neonatal vitamin K prophylaxis in Denmark: Three years’ experience with oral administration during the first three months of life compared with one oral administration at birth. Acta pediatr. 1996;85:1137–1139. [PubMed: 8922069]
  • Haroon Y., Shearer M.J., Rahin S., Gunn W.G., McEnery G., Barkhan P. The content of phylloquinone (vitamin K1) in human milk, cows’ milk and infant formula foods determined by high-performance liquid chromatography. J. Nutr. 1982;112:1105–1117. [PubMed: 7086539]
  • Hart K.T. Study of hydrolysis of urinary metabolites of 2-methyl-1,4-naphthoquinone. Proc. Soc. exp. Biol. Med. 1958;97:848–851. [PubMed: 13554499]
  • Hassan, M.M.A., Mossa, J.S. & Taragan, A.H.U.K. (1988) Analytical profile of phytonadione. In: Florey, K., ed., Analytical Profiles of Drug Substances, New York, Academic Press, Vol. 17, pp. 449–531.
  • Hewson D., Bennett A. Childbirth research data: Medical records or women’s reports? Am. J. Epidemiol. 1987;125:484–491. [PubMed: 3812454]
  • Hill R.J. The uptake of the third oral vitamin K dose in general practice. N.Z. Med. J. 1994;107:177–178. [PubMed: 8177574]
  • Hodges S.J., Bejui J., Leclercq M., Delmas P.D. Detection and measurement of vitamins K1 and K2 in human cortical and trabecular bone. J. Bone Min. Res. 1993;8:1005–1008. [PubMed: 8213250]
  • Hoskin F.C.G., Spinks J.W.T., Jaques L.B. Urinary excretion products of menadione (vitamin K3). Can. J. Biochem. Physiol. 1954;32:240–250. [PubMed: 13150247]
  • Houwen R.H.J., Bouquet J., Bijleveld C.M.A. Bleeding as the first symptom of extra-hepatic biliary atresia. Eur. J. Pediatr. 1987;146:425–426. [PubMed: 3498631]
  • Howe A.M., Webster W.S. Exposure of the pregnant rat to warfarin and vitamin K1: An animal model of intraventricular hemorrhage in the fetus. Teratology. 1990;42:413–420. [PubMed: 2256004]
  • Hwang S.-M. Liquid chromatographic determination of vitamin K1 trans- and cis-isomers in infant formula. J. Assoc. off. anal. Chem. 1985;68:684–689. [PubMed: 4030639]
  • Indyk H.E., Woollard D.C. Vitamin K in milk and infant formulas: Determination and distribution of phylloquinone and menaquinone-4. Analyst. 1997;122:465–469. [PubMed: 9246814]
  • Israels L.G., Walls G.A., Ollmann D.J., Friesen E., Israels E.D. Vitamin K as a regulator of benzo(a)pyrene metabolism, mutagenesis, and carcinogenesis. Studies with rat microsomes and tumorigenesis in mice. J. clin. Invest. 1983;71:1130–1140. [PMC free article: PMC436974] [PubMed: 6304144]
  • Israels L.G., Ollmann D.J., Israels E.D. Vitamin K1 as a modulator of benzo(a)pyrene metabolism as measured by in vitro metabolite formation and in vivo DNA-adduct formation. Int. J. Biochem. 1985;17:1263–1266. [PubMed: 4076526]
  • Israels L.G., Friesen E., Jansen A.H., Israels E.D. Vitamin K1 increases sister chromatid exchange in vitro in human leukocytes and in vivo in fetal sheep cells: A possible role for ‘vitamin K deficiency’ in the fetus. Pediatr. Res. 1987;22:405–408. [PubMed: 3684371]
  • Japan Medical Products Trade Association (1996) Japan Pharmaceutical Reference (JPR). Products and Administration in Japan, 4th Ed., Tokyo, pp. 161–164, 181–186.
  • Jaques L.B., Millar G.J., Spinks J.W.T. The metabolism of the K vitamins. Schweiz. med. Wochenschr. 1954;84:792–796. [PubMed: 13195625]
  • Kameda T., Miyazawa K., Mori Y., Yuasa T., Shiokawa M., Nakamaru Y., Mano H., Hakeda Y., Kameda A., Kumegawa M. Vitamin K2 inhibits osteoclastic bone resorption by inducing osteoclast apoptosis. Biochem. biophys. Res. Commun. 1996;220:515–519. [PubMed: 8607797]
  • Kato T., Watanabe M., Ohta T. Induction of the SOS response and mutations by reactive oxygen-generating compounds in various Escherichia coli mutants defective in the mutM, mutY or soxRS loci. Mutagenesis. 1994;9:245–251. [PubMed: 7934965]
  • Keller C., Matzdorff C., Kemkes-Matthes B. Pharmacology of warfarin and clinical implication. Semin. Thromb. Hemostasis. 1999;25:13–16. [PubMed: 10327215]
  • Klebanoff M.A., Read J.S., Mills J.L., Shiono P.H. The risk of childhood cancer after neonatal exposure to vitamin K. New Engl. J. Med. 1993;329:905–908. [PubMed: 8361503]
  • Kohlmeier M., Salomon A., Saupe J., Shearer M.J. Transport of vitamin K to bone in humans. J. Nutr. 1996;126:1192S–1196S. [PubMed: 8642455]
  • Kossenjans W., Rymaszewski Z., Barankiewicz J., Bobst A., Ashraf M. Menadione-induced oxidative stress in bovine heart microvascular endothelial cells. Microcirculation. 1996;3:39–47. [PubMed: 8846270]
  • Kosuge Y. [Study of developmental pharmacology on vitamin K3. Part 1. Effect of vitamin K3 on the rat fetus.] Folia pharmacol. jpn. 1973;69:285–291. (in Japanese) [PubMed: 4807880]
  • Kowdley K.V., Emond M.J., Sadowski J.A., Kaplan M.M. Plasma vitamin K1 level is decreased in primary biliary cirrhosis. Am. J. Gastroenterol. 1997;92:2059–2061. [PubMed: 9362192]
  • von Kries R. Neonatal vitamin K—Prophylaxis for all. Br. med. J. 1991;303:1083–1084. [PMC free article: PMC1671262] [PubMed: 1747572]
  • von Kries R. Oral versus intramuscular phytomenadione: Safety and efficacy compared. Drug Saf. 1999;21:1–6. [PubMed: 10433349]
  • von Kries R., Hanawa Y. Neonatal vitamin K prophylaxis. Report of Scientific and Standardization Subcommittee on Perinatal Haemostasis. Thromb. Haemostasis. 1993;69:293–295. [PubMed: 8470054]
  • von Kries R., Becker A., Göbel U. Vitamin K in the newborn: Influence of nutritional factors on acarboxy-prothrombin detectability and factor II and VII clotting activity. Eur. J. Pediatr. 1987a;146:123–127. [PubMed: 3569346]
  • von Kries R., Shearer M., McCarthy P.T., Haug M., Harzer G., Göbel U. Vitamin K1 content of maternal milk: Influence of the stage of lactation, lipid composition, and vitamin K1 supplements given to the mother. Pediatr. Res. 1987b;22:513–517. [PubMed: 3684378]
  • von Kries R., Shearer M.J., Gobel U. Vitamin K in infancy. Eur. J. Pediatr. 1988;147:106–112. [PubMed: 3284747]
  • von Kries R., Göbel U., Hachmeister A., Kaletsch U., Michaelis J. Vitamin K and childhood cancer: A population based case–control study in Lower Saxony, Germany. Br. med. J. 1996;313:199–203. [PMC free article: PMC2351611] [PubMed: 8696195]
  • Lamon-Fava S., Sadowski J.A., Davidson K.W., O’Brien M.E., McNamara J.R., Schaefer E.J. Plasma lipoproteins as carriers of phylloquinone (vitamin K1) in humans. Am. J. clin. Nutr. 1998;67:1226–1231. [PubMed: 9625097]
  • LINFO Läkemedelsinformation AB (1998) FASS 1998 Läkemedel i Sverige, Stockholm, pp. 694–695.
  • Lorenz W., Schmal A., Schult H., Lang S., Ohmann C., Weber D., Kapp B., Lüben L., Doenicke A. Histamine release and hypotensive reactions in dogs by solubilizing agents and fatty acids: Analysis of various components in Cremophor EL and development of a compound with reduced toxicity. Agents Actions. 1982;12:64–80. [PubMed: 6177219]
  • Losito R., Owen C.A. Jr, Flock E.V. Metabolism of [14C]menadione. Biochemistry. 1967;6:62–68. [PubMed: 6030337]
  • Losito R., Owen C.A. Jr, Flock E.V. Metabolic studies of vitamin K1-14C and menadione-14C in the normal and hepatectomized rats. Thromb. Diath. Haemorrh. 1968;19:383–388. [PubMed: 4179646]
  • Loughnan P.M., McDougall P.N. Does intramuscular vitamin K1 act as an unintended depot preparation? J. paediatr. Child Health. 1996;32:251–254. [PubMed: 8827545]
  • Mandelbrot L., Guillaumont M., Leclercq M., Lefrère J.J., Gozin D., Daffos F., Forestier F. Placental transfer of vitamin K1 and its implications in fetal hemostasis. Thromb. Haemostasis. 1988;60:39–43. [PubMed: 3187946]
  • Martius, C. (1967) Chemistry and function of vitamin K. In: Seegers W.H., ed., Blood Clotting Enzymology, New York, Academic Press, pp. 551–575.
  • McBurney A., Shearer M.J., Barkhan P. Preparative isolation and characterization of the urinary aglycones of vitamin K1 (phylloquinone) in man. Biochem. Med. 1980;24:250–267. [PubMed: 7306367]
  • McKinney P.A., Juszczak E., Findlay E., Smith K. Case–control study of childhood leukaemia and cancer in Scotland: Findings for neonatal intramuscular vitamin K. Br. med. J. 1998;316:173–177. [PMC free article: PMC2665431] [PubMed: 9468680]
  • McNinch A.W., Upton C., Samuels M., Shearer M.J., McCarthy P., Tripp J.H., Orme R.L.’E. Plasma concentrations after oral or intramuscular vitamin K1 in neonates. Arch. Dis. Child. 1985;60:814–818. [PMC free article: PMC1777471] [PubMed: 4051538]
  • Melgar M.J., Anadon A., Bello J. Effects of menadione on the cardiovascular system. Vet. hum. Toxicol. 1991;33:110–114. [PubMed: 2035237]
  • Menger H., Lin A.E., Toriello H.V., Bernert G., Spranger J.W. Vitamin K deficiency embryopathy: A phenocopy of the warfarin embryopathy due to a disorder of embryonic vitamin K metabolism. Am. J. med. Genet. 1997;72:129–134. [PubMed: 9382132]
  • Meyer T.C., Angus J. The effect of large doses of ‘Synkavit’ in the newborn. Arch. Dis. Child. 1956;31:212–215. [PMC free article: PMC2011962] [PubMed: 13328160]
  • Mezick J. A., Tomkins R.K., Cornwell D.G. Absorption and intestinal lymphatic transport of 14C-menadione. Life Sci. 1968;7:153–158. [PubMed: 5639815]
  • Mirabelli F., Salis A., Marinoni V., Finardi G., Bellomo G., Thor H., Orrenius S. Menadione-induced bleb formation in hepatocytes is associated with the oxidation of thiol groups in actin. Arch. Biochem. Biophys. 1988;264:261–269. [PubMed: 3395123]
  • Morgan W.A. DNA single-strand breakage in mammalian cells induced by redox cycling quinones in the absence of oxidative stress. J. biochem. Toxicol. 1995;10:227–232. [PubMed: 8568837]
  • Morgan W.A., Hartley J.A., Cohen G.M. Quinone-induced DNA single strand breaks in rat hepatocytes and human chronic myelogenous leukaemic K562 cells. Biochem. Pharmacol. 1992;44:215–221. [PubMed: 1642637]
  • Morgan W.A., Kaler B., Bach P.H. The role of reactive oxygen species in adriamycin and menadione-induced glomerular toxicity. Toxicol. Lett. 1998;94:209–215. [PubMed: 9609324]
  • Morrison H., Jernström B., Nordenskjöld M., Thor H., Orrenius S. Induction of DNA damage by menadione (2-methyl-1,4-naphthoquinone) in primary cultures of rat hepatocytes. Biochem. Pharmacol. 1984;33:1763–1769. [PubMed: 6203538]
  • Morrison H., Di Monte D., Nordenskjöld M., Jernström B. Induction of cell damage by menadione and benzo(a)pyrene-3,6-quinone in cultures of adult rat hepatocytes and human fibroblasts. Toxicol. Lett. 1985;28:37–47. [PubMed: 4060194]
  • Motohara K., Matsukura M., Matsuda I., Iribe K., Ikeda T., Kondo Y., Yonekubo A., Yamamoto Y., Tsuchiya F. Severe vitamin K deficiency in breast-fed infants. J. Pediatr. 1984;105:943–945. [PubMed: 6502345]
  • Munday R., Smith B.L., Fowke E.A. Haemolytic activity and nephrotoxicity of 2-hydroxy-1,4-naphthoquinone in rats. J. appl. Toxicol. 1991;11:85–90. [PubMed: 2061555]
  • National Research Council (1989) Recommended Dietary Allowances, 10th Ed., Washington DC, National Academy Press, pp. 107–114.
  • Ngo E.O., Sun T.-P., Chang J.-Y., Wang C.-C., Chi K.-H., Cheng A.-L., Nutter L.M. Menadione-induced DNA damage in a human tumor cell line. Biochem. Pharmacol. 1991;42:1961–1968. [PubMed: 1741774]
  • Nowak D., Chudzik J., Pietras T., Bialasiewicz P. Severe haemorrhagic diathesis in an adult patient with cystic fibrosis after long-term antibiotic treatment of pulmonary infection. Monaldi Arch. Chest Dis. 1997;52:343–345. [PubMed: 9401363]
  • Oakley A., Rajan L., Robertson P. A comparison of different sources of information about pregnancy and childbirth. J. biosoc. Sci. 1990;22:477–487. [PubMed: 2250039]
  • O’Brien D.P., Shearer M.J., Waldron R.P., Horgan P.G., Given H.F. The extent of vitamin K deficiency in patients with cholestatic jaundice: A preliminary communication. J. R. Soc. Med. 1994;87:320–322. [PMC free article: PMC1294558] [PubMed: 8046700]
  • Ochi T. Menadione causes increases in the level of glutathione and in the activity of γ-glutamylcysteine synthetase in cultured Chinese hamster V79 cells. Toxicology. 1996;112:45–55. [PubMed: 8792848]
  • Øie S., Trenk D., Guentert T.W., Mosberg H., Jähnchen E. Disposition of vitamin K1 after intravenous and oral administration to subjects on phenprocoumon therapy. Int. J. Pharm. 1988;48:223–230.
  • Olsen J.H., Hertz H., Blinkenberg K., Verder H. Vitamin K regimens and incidence of childhood cancer in Denmark. Br. med. J. 1994;308:895–896. [PMC free article: PMC2539853] [PubMed: 8173370]
  • Park B.K., Scott A.K., Wilson A.C., Haynes B.P., Breckenridge A.M. Plasma disposition of vitamin K1 in relation to anticoagulant poisoning. Br. J. clin. Pharmacol. 1984;18:655–662. [PMC free article: PMC1463546] [PubMed: 6508974]
  • Parker L., Cole M., Craft A.W., Hey E.N. Neonatal vitamin K administration and childhood cancer in the north of England: Retrospective case–control study. Br. med. J. 1998;316:189–193. [PMC free article: PMC2665412] [PubMed: 9468683]
  • Passmore S.J., Draper G., Brownbill P., Kroll M. Case–control studies of relation between childhood cancer and neonatal vitamin K administration. Br. med. J. 1998a;316:178–184. [PMC free article: PMC2665454] [PubMed: 9468681]
  • Passmore S.J., Draper G., Brownbill P., Kroll M. Ecological studies of relation between hospital policies on neonatal vitamin K administration and subsequent occurrence of childhood cancer. Br. med. J. 1998b;316:184–189. [PMC free article: PMC2665457] [PubMed: 9468682]
  • Pereira S.P., Williams R. Adverse events associated with vitamin K1: Results of a worldwide postmarketing surveillance programme. Pharmacoepidemiol. Drug Saf. 1998;7:173–182. [PubMed: 15073995]
  • Pietersma-de Bruyn A.L.J.M., van Haard P.M.M., Beunis M.H., Hamulyák K., Kuijpers J.C. Vitamin K1 levels and coagulation factors in healthy newborns till 4 weeks after birth. Haemostasis. 1990;20:8–14. [PubMed: 2323682]
  • Pindur G., Mörsdorf S., Schenk J.F., Krischek B., Heinrich W., Wenzel E. The overdosed patient and bleeding with oral anticoagulation. Semin. Thromb. Hemostasis. 1999;25:85–88. [PubMed: 10327226]
  • Pizer B., Boyse J., Hunt L., Mott M. Neonatal vitamin K administration and in vivo somatic mutation. Mutat. Res. 1995;347:135–139. [PubMed: 7565904]
  • Prieto-Alamo M.J., Abril N., Pueyo C. Mutagenesis in Escherichia coli K-12 mutants defective in superoxide dismutase or catalase. Carcinogenesis. 1993;14:237–244. [PubMed: 8382113]
  • Rennie J.M., Kelsall A.W.R. Vitamin K prophylaxis in the newborn—again. Arch. Dis. Child. 1994;70:248–251. [PMC free article: PMC1029755] [PubMed: 8135574]
  • Renzulli P., Tuchschmid P., Eich G., Fanconi S., Schwobel M.G. Early vitamin K deficiency bleeding after maternal phenobarbital intake: Management of massive intracranial haemorrhage by minimal surgical intervention. Eur. J. Pediatr. 1998;157:663–665. [PubMed: 9727852]
  • Rich E.C., Drage C.W. Severe complications of intravenous phytonadione therapy. Postgrad. Med. 1982;72:303–306. [PubMed: 7134079]
  • Richert D.A. Studies on the detoxification of 2-methyl-1,4-naphthoquinone in rabbits. J. biol. Chem. 1951;189:763–768. [PubMed: 14832294]
  • Roman E., Ansell P., Bull D. Leukaemia and non-Hodgkin’s lymphoma in children and young adults: Are prenatal and neonatal factors important determinants of disease? Br. J. Cancer. 1997;76:406–415. [PMC free article: PMC2224068] [PubMed: 9252212]
  • Rote Liste Sekretariat (1998) Rote Liste 1998, Frankfurt, Rote Liste Service GmbH, pp. 84–171, 84–177, 84–180.
  • Royal Pharmaceutical Society of Great Britain (1999) Martindale, The Extra Pharmacopoeia, 13th Ed., London, The Pharmaceutical Press [MicroMedex Online: Health Care Series]
  • Sadtler Research Laboratories (1980) Sadtler Standard Spectra, 1980 Cumulative Index, Philadelphia, PA, p. 881.
  • Sakai A., Miyata N., Takahashi A. Initiating activity of quinones in the two-stage transformation of BALB/3T3 cells. Carcinogenesis. 1995;16:477–481. [PubMed: 7697801]
  • Sanders M.N., Winkelmann R.K. Cutaneous reactions to vitamin K. J. Am. Acad. Dermatol. 1988;19:699–704. [PubMed: 2972759]
  • Sann L., Leclercq M., Frederich A., Bourgeois J., Bethenod M., Bourgeay-Causse M. Pharmacokinetics of vitamin K1 in low-birth-weight neonates. Dev. Pharmacol. Ther. 1985;8:269–279. [PubMed: 4042792]
  • Sawada M., Sofuni T., Hatanaka M., Ishidate M. Jr. Induction of chromosome aberrations in active oxygen-generating systems. 4. Studies with hydrogen peroxide-resistant cells in culture (Abstract No. 52). Mutat. Res. 1987;182:376.
  • Saxena S.P., Fan T., Li M., Israels E.D., Israels L.G. A novel role for vitamin K1 in a tyrosine phosphorylation cascade during chick embryogenesis. J. clin. Invest. 1997;99:602–607. [PMC free article: PMC507841] [PubMed: 9045861]
  • Schardein, J.L. (1993) Chemically Induced Birth Defects, 2nd Ed., New York, Marcel Dekker, p. 552.
  • Schneider D.L., Fluckiger H.B., Manes J.D. Vitamin K1 content of infant formula products. Pediatrics. 1974;53:273–275. [PubMed: 4855823]
  • Schneiderman M.A., Sharma A.K., Mahanama K.R.R., Locke D.C. Determination of vitamin K1 in powdered infant formulas, using supercritical fluid extraction and liquid chromatography with electrochemical detection. J. Assoc. off. anal. Chem. 1988;71:815–817. [PubMed: 3417606]
  • Schubiger G., Tönz O., Grüter J., Shearer M.J. Vitamin K1 concentration in breast-fed neonates after oral or intramuscular administration of a single dose of a new mixed-micellar preparation of phylloquinone. J. pediatr. Gastroenterol. Nutr. 1993;16:435–439. [PubMed: 8315554]
  • Schubiger G., Grüter J., Shearer M.J. Plasma vitamin K1 and PIVKA-II after oral administration of mixed-micellar or Cremophor EL-solubilized preparations of vitamin K1 to normal breast-fed newborns. J. pediatr. Gastroenterol. Nutr. 1997;24:280–284. [PubMed: 9138173]
  • Sharma R.K., Marwaha N., Kumar P., Narang A. Effect of oral water soluble vitamin K on PIVKA-II levels in newborns. Indian Pediatr. 1995;32:863–867. [PubMed: 8635828]
  • Shearer M.J. Vitamin K metabolism and nutriture. Blood Rev. 1992;6:92–104. [PubMed: 1633511]
  • Shearer M.J. Fat-soluble vitamins. Vitamin K. Lancet. 1995;345:229–234. [PubMed: 7823718]
  • Shearer M.J. The roles of vitamins D and K in bone health and osteoporosis prevention. Proc. Nutr. Soc. 1997;56:915–937. [PubMed: 9483660]
  • Shearer M.J., Barkhan P., Webster G.R. 1970 Absorption and excretion of an oral dose of tritiated vitamin K1 in man. Br. J. Haematol. 18, 297–308. [PubMed: 5491582]
  • Shearer M.J., Mallinson C.N., Webster G.R., Barkhan P. Clearance from plasma and excretion in urine, faeces and bile of an intravenous dose of tritiated vitamin K1 in man. Br. J. Haematol. 1972;22:579–588. [PubMed: 5032096]
  • Shearer, M.J., McBurney, A. & Barkhan, P. (1974) Studies on the absorption and metabolism of phylloquinone (vitamin K1) in man. In: Harris, R.S., Munson, P.L., Diczfalusy, E. & Glover, J., Vitamins and Hormones, Advances in Research and Applications, New York, Academic Press, Vol. 32, pp. 513–542.
  • Shearer M.J., Rahm S., Barkhan P., Stimmler L. Plasma vitamin K1 in mothers and their newborn babies. Lancet. 1982;ii:460–463. [PubMed: 6125638]
  • Shearer, M.J., McCarthy, P.T., Crampton, O.E. & Mattock, M.B. (1988) The assessment of human vitamin K status from tissue measurements. In: Suttie J.W., ed., Current Advances in Vitamin K Research, New York, Elsevier, pp. 437–452.
  • Shearer M.J., Bach A., Kohlmeier M. Chemistry, nutritional sources, tissue distribution and metabolism of vitamin K with special reference to bone health. J. Nutr. 1996;126 Suppl. 4:1181S–1186S. [PubMed: 8642453]
  • Shertzer H.G., Låstbom L., Sainsbury M., Moldéus P. Menadione-mediated membrane fluidity alterations and oxidative damage in rat hepatocytes. Biochem. Pharmacol. 1992;43:2135–2142. [PubMed: 1599501]
  • Shinzawa T., Mura T., Tsunei M., Shiraki K. Vitamin K absorption capacity and its association with vitamin K deficiency. Am. J. Dis. Child. 1989;143:686–689. [PubMed: 2729213]
  • Smith P.F., Alberts D.W., Rush G.F. Menadione-induced oxidative stress in hepatocytes isolated from fed and fasted rats: The role of NADPH-regenerating pathways. Toxicol. appl. Pharmacol. 1987;89:190–201. [PubMed: 3603556]
  • Society of Japanese Pharmacopoeia (1996) The Japanese Pharmacopoeia JP XIII, 13th Ed., Tokyo, p. 569.
  • Soedirman J.R., De Bruijn E.A., Maes R.A.A., Hanck A., Grüter J. Pharmacokinetics and tolerance of intravenous and intramuscular phylloquinone (vitamin K1) mixed micelles formulation. Br. J. clin. Pharmacol. 1996;41:517–523. [PMC free article: PMC2042622] [PubMed: 8799516]
  • Solvonuk P.F., Jaques L.B., Leddy J.E., Trevoy L.W., Spinks J.W.T. Experiments with C14-menadione (vitamin K3). Proc. Soc. exp. Biol. Med. 1952;79:597–604. [PubMed: 14920508]
  • Stoeckel K., Joubert P.H., Grüter J. Elimination half-life of vitamin K1 in neonates is longer than is generally assumed: Implications for the prophylaxis of haemorrhagic disease of the newborn. Eur. J. clin. Pharmacol. 1996;49:421–423. [PubMed: 8866641]
  • Sutor A.H., von Kries R., Cornelissen M., McNinch A.W., Andrew M. Vitamin K deficiency bleeding (VKDB) in infancy. Thromb. Haemostasis. 1999;81:456–461. [PubMed: 10102477]
  • Suttie, J.W. (1985) Vitamin K. In: Diplock, A.T., ed., Fat-soluble Vitamins. Their Biochemistry and Applications, Lancaster, PA, Technomic Publishing Co., pp. 225–233, 295–311.
  • Suttie J.W. Recent advances in hepatic vitamin K metabolism and function. Hepatology. 1987;7:367–376. [PubMed: 3549509]
  • Suttie, J.W. (1991) Vitamin K. In: Machlin, L.J., ed., Handbook of Vitamins, 2nd Ed., New York, Marcel Dekker, pp. 145–194.
  • Suttie J.W. The importance of menaquinones in human nutrition. Ann. Rev. Nutr. 1995;15:399–417. [PubMed: 8527227]
  • Swiss Pharmaceutical Society, ed. (1999) Index Nominum, International Drug Directory, 16th Ed., Stuttgart, Medpharm Scientific Publishers [MicroMedex Online: Health Care Series]
  • Taggart W.V., Matschiner J.T. Metabolism of menadione-6,7-3H in the rat. Biochemistry. 1969;8:1141–1146. [PubMed: 4181053]
  • Taylor J.D., Millar G.J., Wood R.J. A comparison of the concentration of C14 in the tissues of pregnant and nonpregnant female rats following the intravenous administration of vitamin K1-C14 and vitamin K3-C14. Can. J. Biochem. Physiol. 1957;35:691–697. [PubMed: 13460789]
  • Thierry M.J., Suttie J.W. Distribution and metabolism of menadiol diphosphate in the rat. J. Nutr. 1969;97:512–516. [PubMed: 5779848]
  • Thijssen H.H.W., Drittij-Reijnders M.J. Vitamin K status in human tissues: Tissue-specific accumulation of phylloquinone and menaquinone-4. Br. J. Nutr. 1996;75:121–127. [PubMed: 8785182]
  • Thomas, J., ed. (1998) Australian Prescription Products Guide, 27th Ed., Victoria, Australian Pharmaceutical Publishing Co., Vol. 1, pp. 687, 1562–1563, 1566, 1570, 1908–1909.
  • Thorp J.A., Gaston L., Caspers D.R., Pal M.L. Current concepts and controversies in the use of vitamin K. Drugs. 1995;49:376–387. [PubMed: 7774512]
  • Tikkanen L., Matsushima T., Natori S., Yoshihira K. Mutagenicity of natural naphthoquinones and benzoquinones in the Salmonella/microsome test. Mutat. Res. 1983;124:25–34. [PubMed: 6355836]
  • Tönz O., Schubiger G. [Neonatal vitamin K prophylaxis and vitamin K deficiency hemorrhage in Switzerland 1986–1988.] Schweiz. med. Wochenschr. 1988;118:1747–1752. (in German) [PubMed: 3206222]
  • Toxopeus C., van Holsteijn I., Thuring J.W.F., Blaauboer B.J., Noordhoek J. Cytotoxicity of menadione and related quinones in freshly isolated rat hepatocytes: Effects on thiol homeostasis and energy charge. Arch. Toxicol. 1993;67:674–679. [PubMed: 8135657]
  • Uchida, K. & Komeno, T. (1988) Relationships between dietary and intestinal vitamin K, clotting factor levels, plasma vitamin K, and urinary Gla. In: Suttie J.W., ed., Current Advances in Vitamin K Research, New York, Elsevier, pp. 477–492.
  • US Pharmacopeial Convention (1994) The 1995 US Pharmacopeia, 23rd Rev./The National Formulary, 18th Rev., Rockville, MD, pp. 946–948, 1224–1226.
  • US Pharmacopeial Convention (1998) USP Dispensing Information, Vol. I, Drug Information for the Health Care Professional, 18th Ed., Rockville, MD, pp. 2984-–986.
  • Usui Y., Tanimura H., Nishimura N., Kobayashi N., Okanoue T., Ozawa K. Vitamin K concentrations in the plasma and liver of surgical patients. Am. J. clin. Nutr. 1990;51:846–852. [PubMed: 2333843]
  • Van Arnum, S.D. (1998) Vitamin K. In: Kroschwitz, J.I. & Howe-Grant, M., eds, Kirk-Othmer Encyclopedia of Chemical Technology, 4th Ed., New York, John Wiley & Sons, Vol. 25, pp. 269–283.
  • Vervoort L.M.T., Ronden J.E., Thijssen H.H.W. The potent antioxidant activity of the vitamin K cycle in microsomal lipid peroxidation. Biochem. Pharmacol. 1997;54:871–876. [PubMed: 9354587]
  • Vest M. Vitamin K in medical practice: Pediatrics. Vitam. Horm. 1966;24:644–663. [PubMed: 5340884]
  • Watanabe K., Sakamoto K., Sasaki T. Comparisons on chemically-induced mutation among four bacterial strains, Salmonella typhimurium TA102 and TA2638, and Escherichia coli WP2/pKM101 and WP2 uvrA/pKM101: Collaborative study II. Mutat. Res. 1998;412:17–31. [PubMed: 9508361]
  • Weber, F. & Rüttimann, A. (1996) Vitamins. 5. Vitamin K. In: Elvers, B. & Hawkins, S., eds, Ullmann’s Encyclopedia of Chemical Technology, 5th Ed., Weinheim, VCH Verlagsgesellschaft, Vol. A27, pp. 488–506.
  • Winterbourn C.C., French J.K., Claridge R.F.C. The reaction of menadione with haemoglobin. Mechanism and effect of superoxide dismutase. Biochem. J. 1979;179:665–673. [PMC free article: PMC1186676] [PubMed: 475774]
  • Woods J.A., Young A.J., Gilmore I.T., Morris A., Bilton R.F. Measurement of menadione-mediated DNA damage in human lymphocytes using the comet assay. Free Radic. Res. 1997;26:113–124. [PubMed: 9257123]
  • Yaguchi M., Miyazawa K., Katagiri T., Nishimaki J., Kizaki M., Tohyama K., Toyama K. Vitamin K2 and its derivatives induce apoptosis in leukemia cells and enhance the effect of all-trans retinoic acid. Leukemia. 1997;11:779–787. [PubMed: 9177427]
  • Yang Y.-M., Simon N., Maertens P., Brigham S., Liu P. Maternal–fetal transport of vitamin K1 and its effect on coagulation in premature infants. J. Pediatr. 1989;115:1009–1013. [PubMed: 2585215]
©International Agency for Research on Cancer, 2000.
Bookshelf ID: NBK401424

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (1.5M)

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...