Paclitaxel and its formulations

https://doi.org/10.1016/S0378-5173(01)00986-3Get rights and content

Abstract

Paclitaxel (Taxol®) is a promising anti-tumor agent with poor water solubility. It is effective for various cancers especially ovarian and breast cancer. Intravenous administration of a current formulation in a non-aqueous vehicle containing Cremophor EL may cause allergic reactions and precipitation on aqueous dilution. Moreover, the extensive clinical use of this drug is somewhat delayed due to the lack of appropriate delivery vehicles. Due to this there is a need for the development of alternate formulation of paclitaxel having good aqueous solubility and at the same time free of any side effects. Various approaches employed so far include cosolvents, emulsions, micelles, liposomes, microspheres nanoparticles, cyclodextrins, pastes, and implants etc. which are discussed in this paper.

Introduction

Paclitaxel, the first of a new class of microtubule stabilizing agents, has been hailed by National Cancer Institute (NCI) as the most significant advance in chemotherapy of the past 15–20 years. Paclitaxel was not a chance discovery but was the outcome of the investigation of over 12 000 natural compounds for anticancer activity (Appendino, 1993)

Paclitaxel is a diterpenoid pseudoalkaloid (Fig. 1) having molecular formula C47H51NO14, corresponding to molecular weight of 853 Da. For anti-tumor activity it is required that entire taxane molecule (Fig. 2) be present, since the ester and the tetraol formed by a low temperature cleavage of paclitaxel are found to be essentially inactive (Wall and Wani, 1996). Paclitaxel was isolated in early 1960s from the bark of Pacific Yew (Taxus brevifolia; family Taxaceae), one of the geographical varieties of yew (Wani et al., 1971). Paclitaxel was obtained in a pure form in 1969 and its structure was published in 1971, after many complexities due to its low concentration and structure complexities (Wani et al., 1971, Bingham, 1994).

The importance of paclitaxel was not recognized until the late 1970s, since, it is difficult to obtain and also due to its low solubility it has a formulation problem. It was in 1979, that Susan Horwitz discovered that paclitaxel has a unique mechanism of action and interest was further stimulated when impressive activity was demonstrated in NCI tumor screening.

Unlike other microtubule agents, such as Vinca alkaloids, which induce the disassembly of microtubules, paclitaxel promotes the polymerization of tubulin (Schiff et al., 1979, Hamel et al., 1981, Parness and Horwitz, 1981, Schiff and Horwitz, 1981, Rowinsky et al., 1990). The microtubule formed in presence of paclitaxel are extraordinarily stable and dysfunctional, thereby causing the death of the cell by disrupting the normal tubule dynamics required for cell division and vital interphase process. Paclitaxel has neoplastic activity particularly against primary epithelial ovarian carcinoma, breast cancer, colon, head, non-small cell lung cancer, and AIDS related Kaposi's sarcoma (Horwitz, 1992, Spencer and Faulds, 1994, Rowinsky and Donehover, 1995, Alshowaier and Nicholls, 1997). This has led to the approval of the drug in many countries for its use as second line treatment of ovarian and breast cancers. In phase II trials with patients treated previously with high dose chemotherapy, the response rate in advanced and refractory ovarian cancer was 30% (Mcguire et al., 1989). The overall response rate in phase I trials in previously treated patients with metastatic breast cancer was 56% (Holmes et al., 1991). In our present paper, we are going to elaborate on the various formulation aspects of paclitaxel, problems associated with the existing formulation and the promising formulation aspects of paclitaxel.

Section snippets

Physical properties and pharmacokinetics

Paclitaxel is white to off-white crystalline powder. It is highly lipophilic, insoluble in water and melts at around 216–217 °C. Its disappearance from plasma is found to be biphasic (Wiernik et al., 1987b). The initial rapid decline represents distribution to the central compartment and elimination of the drug and the later phase is due in part, to the efflux of the drug from the peripheral compartment (Brown et al., 1991). The generally accepted dose is 200–250 mg m−2 and is given as 3 and 24

Limitations

Clinical application of paclitaxel is accompanied by twofold problems.

Paclitaxel dosage form

Paclitaxel is currently formulated in a vehicle composed of 1:1 blend of Cremophor EL (polyethoxylated castor oil) and ethanol which is diluted with 5–20-fold in normal saline or dextrose solution (5%) for administration. This formulation is stable in unopened vials for 5 years at 4 °C. However, lots of problems employing this vehicle have been reported.

(1) One of the substantial problems associated with this formulation is that the ethanol: Cremophor vehicle required to solubilize it is toxic (

Alternative formulations

The primary goal of formulation development for paclitaxel is to eliminate the Cremophor vehicle by reformulation of the drug in a better-tolerated vehicle. Reformulation also provides the possibility of improving the efficacy of paclitaxel based anticancer therapy. A great deal of effort is being directed towards the development of aqueous based formulations for paclitaxel, including soluble semi-synthetic paclitaxel derivatives that do not require solubilisation by Cremophor and that decrease

Conclusion

Paclitaxel (Taxol®), the first taxane in clinical trials, is active against a broad range of cancers that are generally considered to be refractory to conventional therapy. This has lead to the regulatory approval of paclitaxel in the U.S. and many other countries for its use in the palliative therapy of patients with ovarian and breast cancer resistant to chemotherapy. The challenge now is to develop strategies using paclitaxel in the initial therapy of cancer in which cure/improved survival

References (103)

  • W. Mellado et al.

    Preparation and biological activity of taxol acetates

    Biochem. Biophy. Res. Commun.

    (1984)
  • E.S. Park et al.

    Biodegradable polyanhydride devices of cefazolin sodium, bupivacaine, and taxol for local drug delivery: preparation, and kinetics and mechanism of in vitro release

    J. Controlled Release

    (1998)
  • M. Ramaswamy et al.

    Human plasma distribution of free paclitaxel and paclitaxel associated with diblock copolymers

    J. Pharm. Sci.

    (1997)
  • D. Song et al.

    Binding of taxol to plastic glass containers and protein under in vitro conditions

    J. Pharm. Sci.

    (1996)
  • K. Uekama et al.

    Improvement of dissolution and absorption characteristics of benzodiazepines by cyclodextrin complexation

    Int. J. Pharm.

    (1983)
  • M.E. Wall et al.

    Comptothecin and taxol: from discovery to clinic

    J. Ethnopharmacol.

    (1996)
  • J.J. Wheeler et al.

    Polyethylene glycol modified phosoholipids stabilize emulsions prepared from triacylglycerol

    J. Pharm. Sci.

    (1994)
  • J.D. Adams et al.

    Taxol: a history of pharmaceutical development and current pharmaceutical concerns

    J. Nat. Cancer Inst. Monogr.

    (1993)
  • I. Alshowaier et al.

    Management of breast cancer

    Hosp. Pharm.

    (1997)
  • G. Appendino

    Taxol® (paclitaxel): historical and ecological aspects

    Fitoterapia.

    (1993)
  • J.M. Bingham

    Taxus brevifolia: clinical gem or endangered species?

    Aust. J. Hosp. Pharm.

    (1994)
  • M.C. Bissery et al.

    Experimental antitumor activity of taxotere (RP56976, NSC628503) a taxol analogue

    Cancer Res.

    (1991)
  • J.C. Boylan

    Liquids

  • T. Brown et al.

    A phase-I trial of taxol given by 6-hour infusion

    J. Clin. Oncol.

    (1991)
  • T. Cresteil et al.

    Taxol metabolism by human liver microsomes: identification of cytochrome P-450 isozymes involved in its biotransformation

    Cancer Res.

    (1994)
  • H.M. Deutsch et al.

    Synthesis of congeners and prodrugs. 3. Water-soluble prodrugs of taxol with potent antitumor activity

    J. Med. Chem.

    (1989)
  • R.T. Dorr

    Pharmacology and toxicology of Cremophor EL diluent

    Ann. Pharmacother.

    (1994)
  • D. Dye et al.

    Suspected anaphylactic reaction to Cremophor EL

    Br. Med. J.

    (1980)
  • K.S. Etes et al.

    A non-surfactant formulation of alfaxalone based on an amorphous cyclodextrin. Activity studies in rats and dogs

    Int. J. Pharm.

    (1991)
  • G.L. Flynn

    Solubility concepts and their apllication to the formulation of pharmaceutical systems

    J. Parenteral Sci. Technol.

    (1984)
  • D. Friedland et al.

    Hypersenstivity reactions from taxol and etoposide

    J. Natl. Cancer Inst.

    (1993)
  • R.B. Greenwald et al.

    Water soluble taxol 2′-poly (ethylene glycol) ester prodrugs-design and in vivo effectiveness

    J. Med. Chem.

    (1996)
  • E. Hamel et al.

    Interactions of taxol, microtubule associated proteins, and guanine nucleotide in tubulin polymerization

    J. Biol. Chem.

    (1981)
  • F.A. Holmes et al.

    Phase II trials of taxol, an active drug in the treatment of metastatic breast cancer

    Natl. Cancer Inst.

    (1991)
  • R.A. Holton et al.

    First total synthesis of taxol

    J. Am. Chem. Soc.

    (1994)
  • S.B. Horwitz

    Mechanism of action of Taxol

    Trends Pharmacol. Sci.

    (1992)
  • D.L. Howrie et al.

    Anaphylactoid reactions asociated with parenteral cyclosporine use: possible role of Cremophor EL

    Drug Intell. Clin. Pharm.

    (1985)
  • C.A. Jamis-Dow et al.

    Metabolism of taxol by human liver microsomes and effect of inhibitors

    Cancer Res.

    (1993)
  • K. Kataoka et al.

    Block copolymer micelles as vehicle for drug delivery

    J. Controlled Release

    (1993)
  • Kaufman, R.J., Richard, T.J., Fuhrhop, R.W., 1997. Stable O/W emulsion incorporating a taxine (taxol) and method of...
  • R. Klecker et al.

    Effect of cimetidine, probenecid and ketoconazole on the distribution, billiary secretion and metabolism of 3H-taxol in the Sprague–Dawley rat

    Drug metab. Dispos.

    (1994)
  • M. Kongshang et al.

    Interaction of Cremophor EL with human plasma

    J. Biochem.

    (1991)
  • O. Kubo et al.

    Local chemotherapy with slowly-releasing anticancer drug-polymers for malignant brain tumors

    J. Controlled Release

    (1994)
  • G.N. Kumar et al.

    Binding of taxol to human plasma, albumin and acid glycoprotein

    Comm. Chem. Pathol. Pharmacol.

    (1993)
  • Y.W. Lam et al.

    Pharmacokinetics and pharmacodynamics of the taxanes

    J. Oncol. Pharm. Practice.

    (1997)
  • J. Lebmann et al.

    The influence of Cremophor EL on the cell cycle of paclitaxel (Taxol®) in the human cell lines

    Cancer Chemother. Pharmacol.

    (1994)
  • L.L. Lilley et al.

    What you need to know about taxol?

    Am. J. Nurs.

    (1993)
  • S.M. Longnecker et al.

    High performance liquid chrometographic assay for taxol in human plasma and urine and pharmacokinetics in phase I trials

    Cancer Treat. Rep.

    (1987)
  • W. Lorenz et al.

    Histamine release in dogs by Cremophor EL and its derivatives: oxyethylated oleic acid is the most effective constituent

    Agents Actions

    (1977)
  • B.B.J. Lunberg

    A submicron lipid emulsion coated with amphipathic polyethylene glycol for parentral administration of paclitaxel (taxol)

    J. Pharm. Pharmacol.

    (1997)
  • Cited by (982)

    View all citing articles on Scopus
    View full text