Kavallaris et al.
Anticancer therapy with novel tubulin-interacting drugs Maria Kavallaris,1 Nicole M. Verrills,1,2 Bridget T. Hill3 1
Children’s Cancer Institute for Medical Research, Randwick, NSW, Australia,
2
Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW,
Australia, 3 Division de Cancerologie Experimentale, Centre de Recherche Pierre Fabre, Castres, France
Abstract Antimitotic agents that target tubulin, including the taxanes and vinca alkaloids, are important components of current anticancer therapy. Whilst these antimitotic drugs are highly effective in the treatment of a number of cancers, both acquired and intrinsic resistance to these agents is a major clinical problem. Furthermore, the systemic toxicity, and in some cases lack of oral availability, make these agents less than ideal. Recently much effort has been directed on the isolation and synthesis of new antimitotic drugs that target the tubulin/microtubule system and display efficacy against drug-refractory carcinomas. Newly described compounds include structurally diverse natural products, such as dolastatin, epothilones and discodermolide, derivatives and structural analogues of traditional antimitotics, and novel synthetic molecules. Additionally, new developments in drug targeting are improving efficacy and therapeutic indices of traditional agents. A number of promising ‘new generation’ antimitotics are now undergoing clinical testing. These new agents are reviewed here in terms of their mechanism(s) of action on microtubules, effectiveness against drug-resistant tumour cells and clinical C 2002 Elsevier Science Ltd. All rights reserved. potential. °
INTRODUCTION hemotherapeutic agents that target tubulin are important in the treatment of both childhood and adult cancers. The success of these drugs is related to their mechanism of action, that leads to disruption of cell division and induction of apoptosis (Fig. 1). Unfortunately, the clinical usefulness of these agents is limited by the emergence of drug- resistant tumour cells and, in some patients, unacceptably high levels of neurological and bone marrow toxicity. To overcome these obstacles and to improve therapy, investigators in academia and the pharmaceutical industry have focused their efforts on identifying and developing new agents that target tubulin. It is well established that tumours derived from different cells and tissues display a wide variation in their responses to antimitotic drugs, although the reason(s) for this is far from clear. One likely source of tumour-cell differences is that associated with reduced drug accumulation and the multidrug resistance (MDR) phenotype which includes overproduction of the ATP-binding cassette (ABC) transmembrane transporters such as P-glycoprotein (Pgp), the family of multidrug resistance-associated proteins (MRPs)
C
392
c °
2002 Elsevier Science Ltd. All rights reserved.
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
and the breast-cancer resistance protein (BRCP) (reviewed in refs 1–4). Interestingly, whilst Pgp and MRP1 have been shown to have a clear role in resistance to antimitotics,1–3 an elevated expression of BRCP has been associated with sensitivity to vinca alkaloids and to paclitaxel. Another less studied, but important source of tumour-cell differences, is the differential or altered expression of specific microtubule protein family members.5–8 These are likely in turn to affect microtubule dynamics and influence the effects of antimitotic drugs. Moreover, tubulin mutations can affect microtubule stability or interfere with drug-target binding.8–11 Alterations in microtubule structure resulting in altered microtubule dynamics and/or altered binding of antitubulin agents may well constitute a significant mechanism of drug resistance. To date, two of the most clinically useful classes of antimitotic drugs are the vinca alkaloids and the taxanes. Both are from natural products and are used extensively in the treatment of a range of human cancers. The vinca alkaloids (e.g. vincristine, vinblastine and vinorelbine) are cell-cycle specific, blocking cells at the metaphase/anaphase junction of mitosis, and in common with other antimitotic drugs such as colchicine, nocadazole and podophyllotoxin, destabilise microtubules. Taxanes (e.g. paclitaxel and docetaxel) are potent inhibitors of cell proliferation and arrest cells in mitosis, but in contrast to vinca alkaloids, promote the polymerisation of purified tubulin, causing stabilisation and bundling of microtubules.12–14 Both groups of agents target the tubulin/microtubule system. Microtubules are dynamic structures that are constantly growing and shortening and drugs such as the vinca alkaloids and taxanes affect both dynamic instability and treadmilling by interacting with their specific cellular target, β-tubulin.14,15 The clinical success of tubulin as a target for anti-cancer drugs has led the search for new agents that disrupt the tubulin/microtubule system (See Tables 1 and 2). These new agents will be reviewed in terms of their mechanism of action on microtubules, effectiveness against drugresistant tumour cells and clinical potential.
ANTI-MITOTIC DRUGS THAT DESTABILISE MICROTUBULES Vinca alkaloids Vinca alkaloids are natural products isolated from the periwinkle plant Catharanthus roseus. During the past 30 years, the vinca alkaloids vincristine and vinblastine have been used extensively in the treatment of cancer. Innovative chemistry has permitted the semisynthesis of derivatives modified in the velbanamine or ‘upper’ portion of the vinblastine structure.16 From these studies vinorelbine (Navelbine® ) was the first new second-generation vinca alkaloid to emerge and is now used both as a single agent and in combination therapy for cancers as diverse as lung (non small-cell), breast and ovary.17 More recently, a bis-fluorinated vinorelbine derivative, vinflunine, has been synthesised by the application of superacid chemistry.16 Of interest is that whilst all four vinca alkaloids interact with the vinca binding domain on tubulin, there is a clear hierarchy in terms of their overall affinities for purified tubulin. Vinorelbine and, even more so vinflunine, proved weak binders contrasting with the strong binding of vincristine and the
Drug Resistance Updates (2001) 4, 392–401
doi: 10.1054/drup.2002.0230, available online at http://www.idealibrary.com on
New tubulin-targeted cancer treatments
Fig. 1 Effects of tubulin targeting drugs on cancer cells. Tubulin-targeting drugs disrupt microtubule dynamics that are crucial for cell division. Tumour cells that are drug sensitive undergo cell-cycle arrest at the G2/M phase of the cell cycle and subsequently undergo cell death (apoptosis). Resistance to tubulin-targeting drugs can lead to cell survival. Mechanisms responsible for drug resistance include those associated with the multidrug-resistance phenotype (MDR) and alterations in microtubule and associated proteins.
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. Cryptophycins .. . Cryptophycins, isolated from the cyanobacterium Nostoc sp., ... are microtubule-destabilising agents and potent suppressors of ... . microtubule dynamics.25 Their binding to tubulin is competed .
intermediate level of vinblastine. These data suggested that the newer generation compounds may demonstrate reduced neurotoxicity.18,19 More recently, vinorelbine and vinflunine have been shown to affect microtubule dynamics very differently from vinblastine.20 A finding now extended by studying the effects on centromere dynamics which have provided evidence that the specific mechanisms of these three vinca alkaloids are distinct from one another.21 Vinflunine induces cell-cycle arrest in both a time- and concentration-dependent fashion and preclinical evaluations demonstrated that vinflunine displayed superior antitumour activity in vivo (reviewed in refs. 18,19). Evidence has also been presented indicating that the efficacy observed with both vinorelbine and vinflunine is likely to be due to their antivascular effects.18,22,23 Although vinflunine is a substrate for Pgp, the level of cross resistance in MDR-overexpressing cells is much lower than that observed for vinorelbine or vincristine. This correlates with the observation that vinflunine does not induce resistance as readily as vinorelbine following selection under either in vitro or in vivo conditions.24 Overall, vinflunine is a promising new agent that has completed phase I clinical trials and is now undergoing phase II testing. Vinorelbine is also attracting further interest with its oral formulation now in phase I.
c °
by vinca alkaloids. Certain synthetic analogues of cryptophycin, cryptophycin 52 and 55, were identified as potent inducers of Bcl-2 phosphorylation26 and were found to be active in a broad range of murine and human tumour xenografts.27,28 The cryptophycins are active in breast and ovarian tumour cells that overexpress Pgp. In addition to their effectiveness as single agents, they also show great promise in combination with either radiation or with 5-fluorouracil, doxorubicin, paclitaxel or irinotecan.27–29 Cryptophycin 52 is currently in early clinical trials for the treatment of solid tumours. Dolastatin Dolastatin-10 is a novel pentapeptide originally isolated from the marine mollusk Dolabella auricularia. Dolastatin-10 inhibits microtubule assembly and tubulin polymerization and is a non-competitive inhibitor of the binding of vinblastine to tubulin. 30 Although Dolastatin-10 is well tolerated clinically,31 in recent Phase II clinical trials as a single agent it lacked significant activity either in the treatment of hormone refractory prostate cancer32 or at 400 mg/m2 in previously untreated patients with advanced melanoma.33 However, new derivatives of dolastatin-10 such as TZT-1027 have now been identified with superior preclinical activity both in vitro and in vivo.34,35 Their recently reported33,36 anti-vasculature effects and reduced neurotoxicity in animal models further suggest that they may prove valuable new antimicrotubule agents and phase I trials are in progress.
2002 Elsevier Science Ltd. All rights reserved.
Drug Resistance Updates (2001) 4, 392–401
393
Kavallaris et al. Table 1 Microtubule destabilising agents under evaluation Agent Vinca alkaloids Vinflunine Onco-TCS Cryptophycin 52 & 55 Dolastatin Dolastatin-10 TXT-1027 Rhizoxin (NSC 332598) Synthetic Compounds D-24851 A-204197 Ab-conjugated drug C242-DM1
Compound
Derivative of vinflunine Liposomal vincristine Synthetic analogue of Cryptophycin that was originally isolated from cyanobacterium Nostoc sp, Noval pentapeptide originally isolated from marine mollusk Derivative of Dolastatin-10 Novel macrolide antitumor antibiotic
Development stage
Reference
Phase I clinical trials Phase II/III clinical trials for Non Hodgkins Lymphoma Phase I (cryptophysin 52)
19 87, 88 27–29
Phase I/II clinical trials
31–34
Preclinical Phase I/II clinical trials
34–36 97
N-(pyridin-4-yl)-{1-(4-chlorbenzyl)-indol-3-yl}glyoxyl-amid (4-{4-acetyl-4,5-dihydro-5-(3,4,5-trimethoxyphenyl)1,3,4-oxadiazol-2-yl}-N,N-dimethylbenzeneamine)
Preclinical/entering Phase I
37
Preclinical
38
Maytansine-antibody conjugate (tumour activated immunotoxin)
Phase I clinical trials
93
Table 2 Microtubule stabilizing agents under evaluation Agent Taxanes IDN5109 BMS-188797 BMS-184476 BMS-185660 RPR109881A TXD258 (RPR11625A) BMS275183 PG-TXL CT-2103 Polymeric micellar paclitaxel Taxoprexin PTX-DLPC PNU-TXL (PNU166945) Epothilones EpoB dEpoB BMS-247550 (aza-EpoB) dEpoF Discodermolide Laulimalides FR182877 (WS9885B) Synstab A
394
Compound
Development stage
Reference
14β-hydroxy-10-deacetylbaccatin III derivative Semi-synthetic derivative of paclitaxel Ether derivative if paclitaxel Semi-synthetic derivative of paclitaxel Taxane derivative Taxane derivative Novel oral taxane Conjugated poly(L-glutamic acid)-paclitaxel Conjugated poly(L-glutamic acid)-paclitaxel Polymeric (poly(DL-lactide)-block-methoxy polyethylene glycol) micellar paclitaxel DHA-conjugated paclitaxel Aerosol–liposomal paclitaxel Polymer-conjugated paclitaxel
Phase I Clinical Trials PhaseI/II Clinical Trials Phase I/II Clinical Trials Preclinical Phase II Clinical Trials Phase I/II Clinical Trials Phase I/II Clinical Trials Preclinical Phase I Clinical Trials Preclinical
56, 98 52 52 55 51 49, 50 99 100–102 94 103–106
Phase I/II Clinical Trials Preclinical Phase I Clinical trials
91, 92 89 107
Natural product and belongs to 16-membered ring macrolides Epothilone derivative Epothilone derivative Epothilone derivative Polyhydroxylated alketrane
Phase I/II Clinical Trials
58, 59
Derived from Streptomyces sp Small synthetic compound
Drug Resistance Updates (2001) 4, 392–401
c °
2002 Elsevier Science Ltd. All rights reserved.
Final stages of preclinical evaluation Phase I Preclinical Preclinical Preclinical Early preclinical Early preclinical
69, 70, 73 65 73 80–83 77 108, 109 96
New tubulin-targeted cancer treatments .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ANTI-MITOTIC DRUGS THAT STABILISE .. .. MICROTUBULES .. .. .. Taxanes .. Paclitaxel (Taxol® ) was the first natural product described to ... interact with microtubules. Orginally isolated from the bark ... . of the Pacific yew, Taxus brevifolia,40 paclitaxel is a complex . Synthetic compounds Antimitotic compounds that make promising agents for clinical development should have high curative effects in vivo, be effective at destroying multidrug-resistant cells and hopefully have low levels of neurotoxicity. Tubulin-targeted compounds such as the vinca alkaloids and taxanes often cause peripheral neuropathy due to interference of neuronal vesicular transport in axons.6 A recently described microtubule inhibitor, D-24851, appears to fit these criteria. D-24851 or N-(pyridin4-yl)-{1-(4-chlorbenzyl)-indol-3-yl}-glyoxyl-amide is a synthetic compound identified from a cell-based screening assay that destabilises microtubules.37 It was found to be cytotoxic to a range of human tumour cell lines including ovarian (SKOV3), glioblastoma (U87), and pancreatic cancer (ASPC-1). It was also active on tumour cell lines expressing either the Pgp- or MRP-related resistance phenotypes. D-24851 is novel in that it does not compete for binding to either the vincristine- or colchicine-binding sites, implying a distinct tubulin binding profile. Oral D-24851 induced complete tumour regressions in rats bearing Yoshida AH13 sarcoma, markedly superior to the effects obtained with maximal tolerated i.p. doses of either paclitaxel or vincristine.37 Of significance was the absence of any systemic or neuro-toxicity at curative doses of D-24851, although neurotoxicity is difficult to identify in mice. It will be interesting to see whether this molecule with an oral formulation will also have any anti-vascular activity and results of Phase I clinical testing are eagerly awaited. A-204197, a novel oxadiazoline derivative (4-{4-acetyl-4,5dihydro-5-(3,4,5- trimethoxyphenyl)-1,3,4-oxadiazol-2-yl}-N,Ndimethylbenzeneamine), that binds tubulin and causes microtubule depolymerization has recently been reported.38 A-204197 is not a substrate for Pgp, displaying similar antiproliferative properties on cells that either lack, or express Pgp. As with other antimitotic drugs, A-204197 causes cell cycle arrest in G2 -M, increased phosphorylation of G2 -M regulatory proteins and Bcl-2 and induces apoptosis. A-204197 competitively inhibits the binding of 3 H-colchicine but not of {3 H}paclitaxel.38 To date, the number of cell lines examined have been limited and no data for A-204197 in in vivo tumour models have yet been reported. This same group though has now identified indolyloxazoline (A-259745) which maintains all the in vitro activities of the series and showed dose-dependent in vivo activity against the M5076 murine solid tumour, which was unresponsive to vincristine or paclitaxel.39 Again this in vivo activity was reported following administration via the oral route. It still remains to be determined whether this novel compound will be effective in cells resistant to other microtubule agents due to known tubulin/microtubule alterations.6,7 This new series of colchicine-site binders emanating from Abbott Pharmaceuticals are an interesting example of newer antimitotics.
c °
diterpenoid that blocks cell division and induces cell death.41 Paclitaxel-stabilised microtubules were used to elucidate the structure of tubulin42 and the docking analyses that were made possible by this achievement43 increased our understanding of paclitaxel binding and stabilization of microtubules. Paclitaxel binds to the β-tubulin subunit of microtubules at the M-loop, and stabilises lateral contacts between protofilaments,43,44 although the precise conformation of paclitaxel binding remains controversial (for review see45 ). Apart from paclitaxel being an MDR substrate, another limitation is that paclitaxel and its derivative docetaxel are highly hydrophobic, and thus require solubilisation in Cremophor EL® and polysorbate 80 respectively. Some of the toxic reactions observed in patients are thought to be due to these drug vehicles.46–48 The availability of the tubulin:paclitaxel structure has allowed for extensive structure- activity relationship (SAR) analyses and the design of novel taxane derivatives aimed at surmounting some of these limitations. Several such derivatives are in current development by pharamaceutical companies based on their activity against Pgp-expressing tumours. TXD258 is a new taxoid that, like paclitaxel, promotes tubulin assembly in vivo and stabilizes microtubules against cold-induced depolymerization.49 TXD258 has undergone extensive preclinical evaluation and showed excellent tumour regressions and long term survival in xenograft models of pancreas, head and neck, and prostate cancers. Importantly TXD258 is cytotoxic to P-glycoproteinexpressing cells in culture and in a docetaxel-resistant human tumour xenograft model.49 TXD258 also showed antitumour activity in mouse models of intracranial glioblastomas, indicating this agent can cross the blood brain barrier and hence may be useful in the treatment of CNS tumours. Phase I/II clinical trials are in progress with one partial response and a number of minor responses reported.50 Additionally, TXD258 has been shown to be active through oral administration in mouse models, thus circumventing another limitation of the traditional taxanes. RPR109881A is another taxane derivative with a similar mechanism of action to the natural taxanes, shows broad antitumour activity in vitro and in vivo but with minimal recognition by Pgp, and again an ability to cross the blood brain barrier (reviewed in reference 49). In a Phase I clinical trial partial responses were seen in a patient with non-small cell lung cancer and one patient with head and neck cancer, with minor responses in two other patients.51 Various dosing regimes were investigated and Phase II clinical trials are currently underway. Three novel taxane analogues, BMS184476, BMS185660 and BMS188797, have been recently reported on. BMS184476 showed efficacy superior to paclitaxel in three human tumour xenograft models following i.v. administration, with evidence of some activity against paclitaxel-resistant tumours, but not others.52 A therapeutic advantage for BMS188797 over paclitaxel after i.v. administration was also reported in four tumour models, two with partial resistance to paclitaxel.52 It remains to be seen if any therapeutic advantage is to be gained as these newer analogues enter clinical trials. Results of phase I trials reported at ASCO in 200153,54 provided evidence of some partial responses with acceptable toxicity profiles for these two analogues. A water-soluble paclitaxel derivative, BMS185660, with both oral and i.v. activity against human
2002 Elsevier Science Ltd. All rights reserved.
Drug Resistance Updates (2001) 4, 392–401
395
Kavallaris et al. tumour xenografts, has also been reported as a new lead compound in this series.55 Overall though it remains to be established as to whether these newer derivatives have any real therapeutic advantage over paclitaxel or docetaxol. IDN5109 is a new taxane, derived from 14β-hydroxy-10deacetylbaccatin III, selected for its lack of cross-resistance in tumour cell lines expressing the MDR phenotype.56 Encouraging in vivo preclinical data were recently published, indicating that after oral delivery a high level of activity against human ovarian carcinoma xenografts with differing sensitivities to paclitaxel was noted. The good oral bioavailability (48%), coupled with potent antitumour activity makes IDN5109 another attractive candidate for clinical study. Non-taxane microtubule stabilising agents As outlined above, many advances have occurred in the past few years that should improve the therapeutic index of taxanes. Whilst taxanes were the only known compounds to stabilise microtubules for twenty years after their initial discovery, the last decade or so has seen the identification of a number of other, structurally unrelated compounds that appear to act in a taxane-like manner and show great potential as new antimicrotubule chemotherapy agents. These new agents include the natural product epothilones, eleutherobins, sarcodictyins, and their respective analogues, as well as novel synthetic small molecules. Epothilones The 16-member macrolides, epothilone A (EpoA) and epothilone B (EpoB), are natural products first isolated in 1993 from the myxobacterium, Sorangium cellulossum, and initial assessment demonstrated their potency as antifungal agents.57 The epothilones were later shown to have a paclitaxel-like mechanism of action,58 and in contrast to paclitaxel, demonstrated remarkable growth inhibition of MDR cell lines.58–60 Epothilones were the first novel structural class of compounds to be described since the discovery of paclitaxel, that stabilise microtubules. Epothilones were shown to be active in Pgp-expressing and β-tubulin mutated paclitaxel resistant cells, so triggering a surge of investigations to synthesise these molecules. The first solid phase synthesis was described by Nicolaou and colleagues,61 and thus paved the way for the generation of combinatorial libraries containing numerous epothilone analogues. This influx of studies aimed at synthesizing the epothilones has also resulted in the culmination of substantial SAR data.62–65 SAR data point towards a common phamacophore for binding of the epothilones and taxanes.66,67 These compounds competitively inhibit {3 H}paclitaxel binding to microtubules58,59 which, together with data from mutational and pharmacophore analyses, strongly suggests a common binding site.9,67,68 The cytotoxicity of epothilones has been investigated in numerous cancer cell lines,9,58,59,62,64,69,70 with EpoB generally being more cytotoxic than either EpoA or paclitaxel. Most interesting though is the excellent cytotoxicity profiles of epothilones in multidrug resistant cell lines and in a number of paclitaxel resistant cell lines with mutations in β-tubulin.9 Mild resistance to epothilones in culture can be induced by continuous exposure of cultured cells to drug, however this develop396
Drug Resistance Updates (2001) 4, 392–401
c °
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
ment of resistance is far slower than for paclitaxel.64 Analyses of resistant cell lines detected mutations in β-tubulin either in the paclitaxel binding site,68 or in regions that influence microtubule stability,11 in the absence of MDR-1 expression. In in vivo models, EpoA and EpoB had only moderate antitumour activity, attributable to poor metabolic stability, pharmacokinetics and a narrow therapeutic window.70,71 However, results of Phase I trials of EpoB were encouraging and this agent has moved into Phase II clinical testing. BMS-247550 (aza-EpoB) is a semisynthetic EpoB analogue undergoing Phase I clinical trials.65,72 Preclinical studies showed it to be twice as potent as paclitaxel in inducing tubulin polymerization in vitro, and it induced cytotoxicity in tumour cells at low nanomolar concentrations. BMS-247550 retained its antineoplastic activity against human cancers that are naturally insensitive to paclitaxel or those that have developed resistance to paclitaxel, both in vitro and in vivo.71 Two other epothilone analogues which have been studied are 12,13-desoxyepothilones B (dEpoB) and 21-OH-dEpoB (dEpoF). Preclinical evaluation of dEpoB demonstrated it to have superior in vivo antitumour activity than EpoB or paclitaxel, although only small numbers of mice were evaluated.69,70 It was recently reported that dEpoB is at the final preclinical stage undergoing toxicology and pharmacokinetic testing in murine and dog models.73 Although at an earlier stage of development, dEpoF has also shown impressive curative effects in human tumour xenografts in nude mice.73 Eleutherobins, sarcodictyins and laulimalides The natural products eleutherobin, from Eleutherobia sp. and the sarcodicytins A-D, from Sarcodictyon roseium and Eleutherobia aurea, are coral-derived antimitotic agents. Sarcodictyins were originally described as having taxoidlike activity on microtubules, but with much lower relative cytotoxicity.74 The eleutherobins demonstrated similar cell proliferation inhibition to that of paclitaxel, with essentially comparable effects on microtubules.75 Both compounds inhibited the tubulin binding of radiolabelled paclitaxel and proved efficient substrates for Pgp. The successful synthesis of eleutherobin, sarcodictyins A and B, and a variety of analogues76 is now providing sufficient material for further characterisation, and has already provided a methyl ketal sarcodictyin A analogue that shows comparable activity to eleutherobin, yet also exhibiting activity in Pgp-expressing cells. Laulimalide and isolaulimalide are another group of active microtubule-stabilising natural products isolated from a lipophilic extract of the marine sponge Cacospongia mycofijensis, which also maintain their potency in Pgp-expressing cells.77 As the relative antimitotic activity of these agents is generally less than other tubulin stabilising agents in drug sensitive cells, it is unlikely that these agents will progress as anticancer chemotherapeutics. They may however provide the basis for chemical derivatization. Discodermolide Isolated from the marine sponge Discodermia dissoluta, discodermolide, a lactone- bearing polyhydroxylated alkatetraene, was originally realised as a potent immunosuppressor.78,79 Discodermolide inhibits cell proliferation,
2002 Elsevier Science Ltd. All rights reserved.
New tubulin-targeted cancer treatments inducing cell cycle arrest in the G2/M phase, mitotic bundling and affecting microtubule assembly in vivo more rapidly than paclitaxel.80–82 Discodermolide is significantly cytotoxic in Pgp- expressing cells, however not to the extent of the epothilones.82,83 To date, limited supplies from the natural source have hampered in vivo studies. Discodermolide has recently been shown to act synergistically with paclitaxel.83 Furthermore it was found that discodermolide, unlike epothilones and eleutherobin, could not substitute for paclitaxel in a paclitaxel-resistant cell line that requires low concentrations of paclitaxel for normal growth.83 Current progress in the total synthesis of discodermolide.84,85 will hopefully alleviate the supply problem and permit in vivo analysis of this exciting synergism. It will also be interesting to see if structurally altered discodermolides, such as the various acetylated derivatives recently described as showing enhanced antiproliferative activity in the murine leukemia cell line P38886 also exhibit synergistic properties. New delivery for ‘old’ drugs The success of the vinca alkaloids and taxanes is exemplified by their extensive use in the treatment of a wide range of human cancers. As mentioned previously, some clinical drawbacks to treatment with these agents is inadequate delivery of the drug to the tumour cells, the emergence of MDR and neurotoxicity. A number of new formulations for vinca alkaloids and taxanes that aim to overcome these obstacles are now in preclinical or clinical development. One approach has been improved targeting of drugs to tumours using liposomal formulations. Vincristine has been successfully encapsulated into liposomes and is now in phase III clinical trials for the treatment of relapsed non-Hodgkin’s lymphoma and in phase II clinical trials for first line lymphoma, small cell lung cancer and a number of childhood cancers.87,88 Paclitaxel has demonstrated therapeutic potential in the treatment of ovarian, breast and lung cancer.48 However, a number of difficulties have been encountered in the treatment of lung cancer and lung metastasis including reduced active drug accumulation in the tumour due to inactivation of the drug in the blood and liver prior to reaching the tumour. Using a localized delivery system to help overcome this obstacle, a recent preclinical study employed liposome encapsulated paclitaxel and delivered it as an aerosol to mice containing renal cell carcinoma lung metastases.89 A significant reduction in lung weights and visible tumour foci was observed and this also correlated with increased survival in mice inoculated with the tumour cells. Other formulations of paclitaxel include PEG- paclitaxel, dextran conjugated paclitaxel, poly-glu-conjugated paclitaxel, and micellar paclitaxel (reviewed in).90 Another approach to improving the efficacy of taxanes is to enhance the specific targeting of the drug to tumour cells. Anti-tumour agents typically have half-lives far shorter than the cell cycle transit times of most tumour cells. By specifically targeting the tumour the drug concentration will remain high for longer, with more cells to move into cycle, and thus enhanced tumour activity should result. Conjugation of a natural fatty acid to paclitaxel (DHA-paclitaxel or Taxoprexin) has shown superior therapeutic effects in animal models,91,92 with significant increases in the area-under-curve and therapeutic concentration sustainability relative to paclitaxel. Phase I clinc °
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
ical trials with this formulation are in progress in patients with a variety of solid tumours. The idea of conjugating a drug to an antibody that recognises a specific tumour antigen has been around for a long time. Recent progress in the selection of antigens and drug conjugation made the feasibility of this approach more viable. C242-DM1 is a tumour-activated immunotoxin that consists of a humanized antibody, C242, conjugated to a maytansine derivative DM1. Maytansine is a tubulin binding agent and preclinical studies with the antibody-drug conjugate resulted in complete tumour regression in animal models of human pancreatic and non-small cell lung tumours.93 Phase I clinical trials are currently underway using this antibody-tubulin antagonist. If this approach is successful, it could lead to the development of a range of antibody-tubulin antagonist combinations. One advantage, already mentioned, for the newer generation of vinca alkaloids, vinroelbine and vinflunine, is their good oral bioavailability in preclinical in vivo models. Phase I evaluations of oral vinorelbine are underway and similar trials with vinflunine are planned. Other taxane derivatives aimed at improving their solubility and oral bioavailability are also in clinical trials. The conjugation of a poly (L-glutamic acid) to paclitaxel creates a water-soluble molecule that is currently being trialed.94 The ability to dispense with Cremophor EL® and ethanol vehicles is predicted to achieve a significant improvement in the pharmocokinetics of these taxanes. BMS275183 is a novel oral taxane that has good oral availability in rats and dogs, and preliminary Phase I clinical data indicate effective bioavailability in humans.95 Small molecule drugs The tubulin/microtubule system still remains one of the most important targets for cancer design. The search for small molecules as drugs that disrupt cell division led Haggarty and colleagues to utilise a high-throughput phenotype-based screen for identifying cell-permeable small molecules that affect mitosis.96 Of the 139 compounds found to increase the amount of phosphorylated nucleolin, indicating likely mitotic arrest, in asynchronous A549 lung epithelial tumour cells, 52 compounds were shown to destabilize microtubules, and one compound to stabilize microtubules. This compound, designated Synstab A, stabilised purified microtubules through a direct interaction with tubulin in a similar manner to paclitaxel, and led to microtubule bundles in interphase, disrupted spindles and caused abnormal mitotic chromosome distribution in kidney epithelial cells. The observed effects were reversible and Synstab A partially displaced fluorescently labeled paclitaxel, indicating a potential overlap in the tubulin binding site. The structure of this molecule is distinct from previously described microtubule stabilising agents, and highlights the potential for phenotype-based screening to identify novel anti-microtubule agents. This same approach is now being utilized by a number of pharmaceutical companies and offers the potential for identifying a number of novel antimitotics from natural product sources and chemical libraries. CONCLUSIONS AND FUTURE PERSPECTIVE In recent years the enormous effort invested in identifying new antimitotic therapies that target the tubulin/microtubule system has yielded a number of promising drugs for the treatment
2002 Elsevier Science Ltd. All rights reserved.
Drug Resistance Updates (2001) 4, 392–401
397
Kavallaris et al. of advanced stage cancers (Tables 1 and 2). In some cases, these new or modified anti-microtubule agents inhibit the growth of tumour cells that are MDR, and/or resistant tumour cells with paclitaxel-induced β-tubulin mutations. The ability of many of these agents to by-pass resistance associated with tubulin/microtubule changes including tubulin mutations, altered expression of tubulins and microtubule-associated proteins, still remains to be determined. An important parameter in evaluating any new anti-cancer agent is its therapeutic index, whereby the drug selectively kills tumour cells in the clinical setting. New agents that are highly effective in vivo may fail Phase I trials due to unacceptable toxicities. Many of the newer agents discussed in this review are now in early Phase I/II clinical trials. Whilst preliminary results for a number of these new tubulin targeted agents are encouraging, it is likely that the true potential of certain of these drugs will be not as single agents, but rather in combination therapies.110,111 In this respect, the various studies identifying synergistic interactions between these newer agents and ‘standard’ cytotoxics are encouraging. Combinations of agents with differential effects on the same overall target, namely the tubulin/microtubule system, may also prove effective in our search for active therapies to use against drug refractory cancers.
Received 18 January 2002; Revised and Accepted 4 February 2002 Correspondence to: Dr Maria Kavallaris, Children’s Cancer Institute Australia for Medical Research, High Street (PO Box 81), Randwick, NSW, Australia 2031, Tel: 61-2-9382 1823; Fax: 61-2-9382 1850; E-mail:
[email protected]
References 1. Gottesman MM, Pastan I. Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem 1993; 62: 385–427. 2. Kavallaris M. The role of Multidrug Resistance-associated Protein (MRP) gene expression in drug resistance. Anti-Cancer Drugs 1997; 8: 17–25. 3. Borst P, Evers R, Kool M et al. A family of drug transporters: the multidrug resistance-associated proteins. J Natl Cancer Inst 2000; 92: 1295–1302. 4. Ross DD, Yang W, Abruzzo LV et al. Atypical multidrug resistance: Breast cancer resistance protein messenger RNA expression in mitoxantrone-selected cell lines. J Natl Cancer Inst 1999; 91: 429–433. 5. Kavallaris M, Kuo DY-S, Burkhart CA et al. Taxol-resistant epithelial ovarian tumors are associated with altered expression of specific β-tubulin isotypes. J Clin Invest 1997; 100: 1282–1293. 6. Cabral F. Factors determining cellular mechanisms of resistance to antimitotic drugs. Drug Resist Updates 2001; 4: 3–8. 7. Burkhart CA, Kavallaris M, Horwitz SB. The role of β-tubulin isotypes in resistance to antimitotic drugs. Biochim Biophys Acta 2001; 1471: 1–9. 8. Kavallaris M, Tait AS, Walsh BJ et al. Multiple microtubule alterations are associated with vinca alkaloid resistance in human leukaemia cells. Cancer Res 2001; 61: 5803–5809.
398
Drug Resistance Updates (2001) 4, 392–401
c °
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
9. Giannakakou P, Sackett DL, Kang YK et al. Paclitaxel-resistant human ovarian cancer cells have mutant beta-tubulins that exhibit impaired paclitaxel-driven polymerization. J Biol Chem 1997; 272: 17118–17125. 10. Gonzalez-Garay ML, Chang L, Blade K et al. A β-tubulin leucine cluster involved in microtubule assembly and paclitaxel resistance. J Biol Chem 1999; 274: 23875–23882. 11. He L, Yang C-PH, Horwitz SB. Mutations in β-tubulin map to domains involved in regulation of microtubule stability in epothilone-resistant cell lines. Mol Cancer Therapeut 2001; 1: 3–10. 12. Schiff PB, Horwitz SB. Taxol stabilizes microtubules in mouse fibroblast cells. Proc Natl Acad Sci USA 1980; 77: 1561–1565. 13. Jordan MA, Toso RJ, Thrower D et al. Mechanism of mitotic block and inhibition of cell proliferation by taxol at low concentrations. Proc Natl Acad Sci USA 1993; 90: 9552–9556. 14. Derry WB, Wilson L, Jordan MA. Substoichiometric binding of taxol suppresses microtubule dynamics. Biochemistry 1995; 34: 2203–2211. 15. Ngan VK, Bellman K, Panda D et al. Novel actions of the antitumor drugs vinflunine and vinorelbine on microtubules. Cancer Res 2000; 60: 5045–5051. 16. Fahy J. Modifications in the “upper” velbenamine part of the vinca alkaloids have major implications for tubulin interacting activities. Curr Pharm Des 2001; 7: 1181–1197. 17. Rowinsky EK, Donehower RC. Antimicrotubule Agents. In: VT Devita, S Hellman, SA. Rosenberg (eds.) Cancer Principles and Practice of Oncology, pp. 467–483. Philadelphia: Lippincott-Raven, 1997. 18. Kruczynski A, Hill BT. Vinflunine, the latest vinca alkaloid in clinical development. A review of its preclinical anticancer properties. Crit Rev Oncol Hematol 2001; 40: 159–173. 19. Hill BT. Vinflunine, a second generation novel vinca alkaloid with a distinctive pharmacological profile, now in clinical development and prospects for future mitotic blockers. Curr Pharm Des 2001; 7: 1199–1212. 20. Ngan VK, Bellman K, Hill BT et al. Mechanism of mitotic block and inhibition of cell proliferation by the semisynthetic vinca alkaloids vinorelbine and its newer derivative vinflunine. Mol Pharmacol 2001; 60: 225–232. 21. Okouneva T, Wilson L, Hill B et al. How do the newer vinca alkaloids vinflunine and vinorelbine block mitosis? Effects on centromere movements in living mitotic U20s cells. Proc Am Assoc Cancer Res 2002; 43: in press. 22. Holwell SE, Hill BT, Bibby MC. Anti-vascular effects of vinflunine in the MAC 15A transplantable adenocarcinoma model. Br J Cancer 2001; 84: 290–295. 23. Kruczynski A, Ricome C, Astruc J et al. Significant antiangiogenic activity and marked inhibition of growth of an orthotopic colon cancer model and colon cancer liver metastases by vinflunine, the most recent vinca alkaloid in clinical development. Proc Am Assoc Cancer Res 2002; 43: in press. 24. Eti´evant C, Kruczynski A, Tait AS et al. Markedly diminished drug-resistance inducing properties of vinflunine (200 ,200 difluoro-30 ,40 -dihydrovinorelbine) relative to vinorelbine, identified in murine and human tumour cells in vivo and in vitro, with clinical implications. Cancer Chemother Pharmacol 2001; 48: 62–70. 25. Panda D, DeLuca K, Williams D et al. Antiproliferative mechanisms of action of cryptophycin-52:Kinetic stabilization of microtubule
2002 Elsevier Science Ltd. All rights reserved.
New tubulin-targeted cancer treatments
26.
27.
28.
29. 30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41. 42. 43. 44.
dynamics by high-affinity binding to microtubule ends. Proc Natl Acad Sci USA 1998; 95: 9313–9318. Lu K, Dempsey J, Schultz RM et al. Cryptophycin-induced hyperphosphorylation of Bcl-2, cell cycle arrest and growth inhibition in human H460 NSCLC cells. Cancer Chemother Pharmacol 2001; 47: 170–178. Teicher BA, Forler P, Menon K et al. Cryptophycin 52 and cryptophycin 55 in sequential and simultaneous combination treatment regimens in human tumour xenografts. In Vivo 2000; 14: 471–480. Menon K, Alvarez E, Forler P et al. Antitumour activity of cryptophycins: effect of infusion time and combination studies. Cancer Chemother Pharmacol 2000; 46: 142–149. Shih C, Teicher BA. Cryptophycins: A novel class of potent antitumor depsipeptides. Curr Pharm Des 2001; 7: 1259–1276. Bai R, Taylor GF, Schmidt JM et al. Interaction of dolastatin 10 with tubulin: induction of aggregation and binding and dissociation reactions. Mol Pharmacol 1995; 47: 965–976. Pitot HC, McElroy EAJ, Reid JM et al. Phase I trial of dolastatin-10 (NSC 376128) in patients with advanced solid tumors. Clin Cancer Res 1999; 5: 525–531. Vaishampayan U, Glode M, Du W et al. Phase II study of dolastatin-10 in patients with hormone-refractory metastatic prostate adenocarcinoma. Clin Cancer Res 2000; 6: 4205–4208. Margolin K, Longmate J, Synold TW et al. Dolastatin-10 in metastatic melanoma: a phase II and pharmokinetic trial of the California Cancer Consortium. Invest New Drugs 2002; 19: 335–340. Otani M, Natsume T, Watanabe JI et al. TZT-1027, an antimicrotubule agent, attacks tumor vasculature and induces tumor cell death. Jpn J Cancer Res 2000; 91: 837–844. Natsume T, Watanabe J, Tamaoki S et al. Characterization of the interaction of TZT-1027, a potent antitumor agent, with tubulin. Jpn J Cancer Res 2000; 91: 737–747. Ogawa T, Mimura Y, Isowa K et al. An antimicrotubule agent, TZT-1027, does not induce neuropathologic alterations which are detected after administration of vincristine or paclitaxel in animal models. Toxicol Lett 2001; 121: 97–106. Bacher G, Nickel B, Emig P et al. D-24851, a novel synthetic microtubule inhibitor, exerts curative antitumoral activity in vivo, shows efficacy toward multidrug-resistant tumor cells, and lacks neurotoxicity. Cancer Res 2001; 61: 392–399. Tahir SK, Han EK, Credo B et al. A-204197, a new tubulin-binding agent with antimitotic activity in tumor cell lines resistant to known microtubule inhibitors. Cancer Res 2001; 61: 5480–5485. Szczepankiewicz BG, Liu G, Jae H-S et al. New antimitotic agents with activity in multi-drug resistant cell lines and in vivo efficacy in murine tumor models. J Med Chem 2001; 44: 4416–4430. Wani MC, Taylor HL, Wall ME et al. Plant antitumor agents VI. The isolation and structure of taxol, a novel antileukemic and antitumour agent from taxus brevifolia. J Am Chem Soc 1971; 93: 2325–2327. Horwitz SB. Mechanism of action of taxol. Trends Pharmacol Sci 1992; 13: 134–136. Nogales E, Wolf SG, Downing KH. Structure of the αβ tubulin dimer by electron crystallography. Nature 1998; 391: 199–203. Nogales E, Whittaker M, Milligan RA et al. High resolution model of the microtubule. Cell 1999; 96: 79–88. Amos LA, Lowe J. How Taxol stabilises microtubule structure. Chem Biol 1999; 6: R65–69.
c °
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
45. Downing KH. Structural basis for the interaction of tubulin with proteins and drugs that affect microtubule stability. Annu Rev Cell Dev Biol 2000; 16: 89–111. 46. Dorr RT. Pharmacology and toxicology of cremophor EL diluent. Ann Pharmacother 1994; 28: 511–514. 47. van Oosterom AT. Docetaxel (Taxotere): an effective agent in the management of second-line breast cancer. Semin Oncol 1995; 22: 22–28. 48. Rowinsky EK. The development and clinical utility of the taxane class of antimicrotubule chemotherapy agents. Annu Rev Med 1997; 48: 353–374. 49. Bissery MC. Preclinical evaluation of new taxoids. Curr Pharm Des 2001; 7: 1251–1257. 50. Goetz AD, Denis LJ, Rowinsky EK et al. Phase I and Pharmacokinetic Study of RPR116258A, a Novel Taxane Derivative, Administered Intravenously over 1 Hour Every 3 Weeks. Proc Am Soc Clin Oncol 2001; 20: 106a (abstract 419). 51. Kurata T, Shimada Y, Tamura T et al. Phase I and pharmacokinetic study of a new taxoid, RPR 109881A, given as a 1-hour intravenous infusion in patients with advanced solid tumors. J Clin Oncol 2000; 18: 3164–3171. 52. Rose WC, Fairchild C, Lee FYF. Preclinical antitumor activity of two novel taxanes. Cancer Chemother Pharmacol 2001; 97–105. 53. Beeram M, Hidalgo M, Rodriguez G et al. A phase I and pharmacokinetic (PK) study of the novel taxane BMS 184476 administered As a 1-hour intravenous (IV) infusion weekly. Proc Am Soc Clin Oncol 2001; 20: 106a (abstract 421). 54. Goldstein LJ, Vaders L, Rogatko A et al. Phase I Study of BMS-188797, a New Taxane Analog, Given Weekly in Patients with Advanced Malignancies. Proc Am Soc Clin Oncol 2001; 20: 86b (abstract 2093). 55. Rose WC, Lee FY, Golik J et al. Preclinical oral antitumor activity of BMS-185660, a paclitaxel derivative. Cancer Chemother Pharmacol 2000; 46: 246–250. 56. Nicoletti MI, Colombo T, Rossi C et al. IDN5109, a taxane with oral bioavailability and potent antitumor activity. Cancer Res 2000; 60: 842–846. 57. Gerth K, Bedorf N, Hofle G et al. Epothilons A and B: antifungal and cytotoxic compounds from Sorangium cellulosum (Myxobacteria). Production, physico-chemical and biological properties. J Antibiot 1996; 49: 560–563. 58. Bollag DM, McQueney PA, Zhu J et al. Epothilones, a new class of microtubule-stabilizing agents with a taxol-like mechanism of action, Cancer Res 1995; 55: 2325–2333. 59. Kowalski RJ, Giannakakou P, Hamel E. Activities of the microtubule-stabilizing agents epothilones A and B with purified tubulin and in cells resistant to paclitaxel (Taxol(R)). J Biol Chem 1997; 272: 2534–2541. 60. Muhlradt PF, Sasse F. Epothilone B stabilizes microtubuli of macrophages like taxol without showing taxol-like endotoxin activity. Cancer Res 1997; 57: 3344–3346. 61. Nicolaou KC, Winssinger N, Pastor J et al. Synthesis of epothilones A and B in solid and solution phase. Nature 1997; 387: 268–272. 62. Nicolaou KC, Finlay MR, Ninkovic S et al. Synthesis and biological properties of C12,13-cyclopropyl-epothilone A and related epothilones. Chem Biol 1998; 5: 365–372. 63. Mulzer J, Mantoulidis A, Ohler E. Total syntheses of epothilones B and D. J Org Chem 2000; 65: 7456–7467. 64. Altmann KH, Wartmann M, O’Reilly T. Epothilones and related structures–a new class of microtubule inhibitors with potent in
2002 Elsevier Science Ltd. All rights reserved.
Drug Resistance Updates (2001) 4, 392–401
399
Kavallaris et al.
65.
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
79. 80.
81.
400
vivo antitumor activity. Biochim Biophys Acta 2000; 1470: M79–91. Stachel SJ, Chappell MD, Lee CB et al. On the total synthesis and preliminary biological evaluations of 15(R) and 15(S) aza-dEpoB: a Mitsunobu inversion at C15 in pre-epothilone fragments. Org Lett 2000; 2: 1637–1639. Ojima I, Chakravarty S, Inoue T et al. A common pharmacophore for cytotoxic natural products that stabilize microtubules. Proc Natl Acad Sci USA 1999; 96: 4256–4261. He L, Jagtap PG, Kingston DGI et al. A common pharmacophore for Taxol and the epothilones based on the biological activity of a taxane molecule lacking a C-13 side chain. Biochemistry 2000; 39: 3972–3978. Giannakakou P, Gussio R, Nogales E et al. A common pharmacophore for epothilone and taxanes: Molecular basis for drug resistance conferred by tubulin mutations in human cancer cells. Proc Natl Acad Sci USA 2000; 97: 2904–2909. Chou TC, Zhang XG, Balog A et al. Desoxyepothilone B: an efficacious microtubule-targeted antitumor agent with a promising in vivo profile relative to epothilone B. Proc Natl Acad Sci USA 1998; 95: 9642–9647. Chou TC, Zhang XG, Harris CR et al. Desoxyepothilone B is curative against human tumor xenografts that are refractory to paclitaxel. Proc Natl Acad Sci USA 1998; 95: 15798–15802. Lee FY, Borzilleri R, Fairchild CR et al. BMS-247550: a novel epothilone analog with a mode of action similar to paclitaxel but possessing superior antitumor efficacy. Clin Cancer Res 2001; 7: 1429–1437. Colevas AD, West PJ, Cheson BD. Clinical trials referral resource. Current clinical trials of epothilone B analog (BMS-247550). Oncology 2001; 15: 1168–1169. Chou TC, O’Connor OA, Tong WP et al. The synthesis, discovery, and development of a highly promising class of microtubule stabilization agents: curative effects of desoxyepothilones B and F against human tumor xenografts in nude mice. Proc Natl Acad Sci USA 2001; 98: 8113–8118. Hamel E, Sackett DL, Vourloumis D et al. The coral-derived natural products eleutherobin and sarcodictyins A and B: effects on the assembly of purified tubulin with and without microtubuleassociated proteins and binding at the polymer taxoid site. Biochemistry 1999; 38: 5490–5498. Long BH, Carboni JM, Wasserman AJ et al. Eleutherobin, a novel cytotoxic agent that induces tubulin polymerization, is similar to paclitaxel (Taxol). Cancer Res 1998; 58: 1111–1115. Nicolaou KC, Pfefferkorn J, Xu J et al. Total synthesis and chemical biology of the sarcodictyins, Chem Pharm Bull 1999; 47: 1199–1213. Mooberry SL, Tien G, Hernandez AH et al. Laulimalide and isolaulimalide, new paclitaxel-like microtubule-stabilizing agents, Cancer Res 1999; 59: 653–660. Longley RE, Caddigan D, Harmody D et al. Discodermolide–a new, marine-derived immunosuppressive compound. I. In vitro studies. Transplantation 1991; 52: 650–656. Longley RE, Gunasekera SP, Faherty D et al. Immunosuppression by discodermolide. Ann N Y Acad Sci 1993; 696: 94–107. ter Haar E, Kowalski RJ, Hamel E et al. Discodermolide, a cytotoxic marine agent that stabilizes microtubules more potently than taxol. Biochemistry 1996; 35: 243–250. Hung DT, Chen J, Schreiber SL. (+)-Discodermolide binds to microtubules in stoichiometric ratio to tubulin dimers, blocks taxol
Drug Resistance Updates (2001) 4, 392–401
c °
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
82.
83.
84. 85.
86.
87.
88. 89.
90.
91.
92.
93. 94.
95.
96.
97.
98.
2002 Elsevier Science Ltd. All rights reserved.
binding and results in mitotic arrest. Chem Biol 1996; 3: 287–293. Kowalski RJ, Giannakakou P, Gunasekera SP et al. The microtubule-stabilizing agent discodermolide competitively inhibits the binding of paclitaxel (Taxol) to tubulin polymers, enhances tubulin nucleation reactions more potently than paclitaxel, and inhibits the growth of paclitaxel-resistant cells. Mol Pharmacol 1997; 52: 613–622. Martello LA, McDaid HM, Regl DL et al. Taxol and discodermolide represent a synergistic drug combination in human carcinoma cell lines. Clin Cancer Res 2000; 6: 1978–1987. Smith AB, 3rd, Kaufman MD, Beauchamp TJ et al. Gram-scale synthesis of (+)-discodermolide. Org Lett 1999; 1: 1823–1826. Paterson I, Florence GJ, Gerlach K et al. A practical synthesis of (+)-discodermolide and analogues: fragment union by complex aldol reactions. J Am Chem Soc 2001; 123: 9535–9544. Gunasekera SP, Longley RE, Isbrucker RA. Acetylated analogues of the microtubule-stabilizing agent discodermolide: preparation and biological activity. J Nat Prod 2001; 64: 171–174. Sarris AH, Hagemeister F, Romaguera J et al. Liposomal vincristine in relapsed non-Hodgkin’s lymphomas: early results of an ongoing phase II trial. Ann Oncol 2000; 11: 69–72. Gelmon KA, Tolcher A, Diab AR et al. Phase I study of liposomal vincristine. J Clin Oncol 1999; 17: 697–705. Koshkina NV, Waldrep JC, Roberts LE et al. Paclitaxel liposome aerosol treatment induces inhibition of pulmonary metastases in murine renal carcinoma model. Clin Cancer Res 2001; 7: 3258–3262. Wood KW, Cornwell WD, Jackson JR. Past and future of the mitotic spindle as an oncology target. Curr Opin Pharmacol 2001; 1: 370–377. Bradley MO, Webb NL, Anthony FH et al. Tumor targeting by covalent conjugation of a natural fatty acid to paclitaxel. Clin Cancer Res 2001; 7: 3229–3238. Bradley MO, Swindell CS, Anthony FH et al. Tumor targeting by conjugation of DHA to paclitaxel. J Control Rel 2001; 74: 233–236. Smith S. Technology evaluation: C242-DM1, ImmunoGen Inc. Curr Opin Mol Ther 2001; 3: 198–203. Todd R, Sludden J, Boddy AV, Griffin MJ, Robson L, Cassidy J, Bissett D, Main M, Brannan MD, Elliott S, Fishwick K, Verrill M, Calvert H. Phase I and Pharmacological Study of CT-2103, a Poly (L-glutamic Acid)-Paclitaxel Conjugate. Proc Am Soc Clin Oncol 2001; 20: 111a (abstract 439). Trump DL, Egorin M, Ramanathan R et al. A Novel Oral Taxane (BMS-275183): a Phase I Trial with Evaluation of Pharmacokinetics, Pharmacodynamics and Bioavailability. Proc Am Soc Clin Oncol 2001; 20: 109a (abstract 433). Haggarty SJ, Mayer TU, Miyamoto DT et al. Dissecting cellular processes using small molecules: identification of colchicine-like, taxol-like and other small molecules that perturb mitosis. Chem Biol 2000; 7: 275–286. Tolcher AW, Aylesworth C, Rizzo J et al. A phase I study of rhizoxin (NSC 332598) by 72-hour continuous intravenous infusion in patients with advanced solid tumors. Ann Oncol 2000; 11: 333–338. Ojima I, Slater JC, Michaud E et al. Syntheses and structure-activity relationships of the second-generation antitumor taxoids: exceptional activity against drug-resistant cancer cells. J Med Chem 1996; 39: 3889–3896.
New tubulin-targeted cancer treatments 99. Rose WC, Long BH, Fairchild CR et al. Preclinical pharmacology of BMS-275183, an orally active taxane. Clin Cancer Res 2001; 7: 2016–2021. 100. Li C, Yu DF, Newman RA et al. Complete regression of well-established tumors using a novel water-soluble poly(L-glutamic acid)-paclitaxel conjugate. Cancer Res 1998; 58: 2404–2409. 101. Li C, Newman RA, Wu QP et al. Biodistribution of paclitaxel and poly(L-glutamic acid)-paclitaxel conjugate in mice with ovarian OCa-1 tumor. Cancer Chemother Pharmacol 2000; 46: 416–422. 102. Oldham EA, Li C, Ke S et al. Comparison of action of paclitaxel and poly(L-glutamic acid)-paclitaxel conjugate in human breast cancer cell. Int J Oncol 2000; 16: 125–132. 103. Zhang X, Burt HM, Von Hoff D et al. An investigation of the antitumour activity and biodistribution of polymeric micellar paclitaxel. Cancer Chemother Pharmacol 1997; 40: 81–86. 104. Zhang X, Burt HM, Mangold G et al. Anti-tumor efficacy and biodistribution of intravenous polymeric micellar paclitaxel. Anticancer Drugs 1997; 8: 696–701. 105. Ramaswamy M, Zhang X, Burt HM et al. Human plasma distribution of free paclitaxel and paclitaxel associated with diblock copolymers, J Pharm Sci 1997; 86: 460–464.
c °
.. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..
106. Leung SY, Jackson J, Miyake H et al. Polymeric micellar paclitaxel phosphorylates Bcl-2 and induces apoptotic regression of androgen-independent LNCaP prostate tumors. Prostate 2000; 44: 156–163. 107. Meerum Terwogt JM, ten Bokkel Huinink WW, Schellens JH et al. Phase I clinical and pharmacokinetic study of PNU166945, a novel water-soluble polymer-conjugated prodrug of paclitaxel. Anticancer Drugs 2001; 12: 315–323. 108. Sato B, Nakajima H, Hori Y et al. A new antimitotic substance, FR182877. II. The mechanism of action. J Antibiot 2000; 53: 204–206. 109. Sato B, Muramatsu H, Miyauchi M et al. A new antimitotic substance, FR182877. I. Taxonomy, fermentation, isolation, physico-chemical properties and biological activities. J Antibiot 2000; 53: 123–130. 110. Shah MA, Schwartz GK. The relevance of drug sequence in combination chemotherapy. Drug Resist Updates 2000; 3: 335–356. 111. Sparreboom A, Nooter K. Does P-glycoprotein play a role in anticancer drug pharmacokinetics? Drug Resist Updates 2000; 3: 357–363.
2002 Elsevier Science Ltd. All rights reserved.
Drug Resistance Updates (2001) 4, 392–401
401