8
Pediatr Blood Cancer 2009;53:1180–1187 REVIEW Vincristine: Can Its Therapeutic Index Be Enhanced? Andrew Moore, MBBS 1 * and Ross Pinkerton, MD, FRCPCH 2 INTRODUCTION Although first used successfully in adults and children as a cytotoxic agent in 1962, vincristine has been known for its medicinal properties since the 17th century. Extracts from the plant Vinca rosea have been described in medicinal folklore as being effective for the treatment of haemorrhage, scurvy, toothache, wound healing, diabetic ulcers and hyperglycaemia [1]. Vincristine exerts its cytotoxic effects via interference with microtubule formation and mitotic spindle dynamics, disruption of intracellular transport and decreased tumour blood flow, with the latter probably as a consequence of anti-angiogenesis [1,2]. The ultimate result of vincristine-mediated cellular disruption is apoptosis [1]. The most frequent and clinically important side-effect of vincristine is neurotoxicity, characteristically resulting in an autonomic and peripheral sensory-motor polyneuropathy. Central nervous system toxicity is uncommon and can include the syndrome of inappropriate anti-diuretic hormone (SIADH) secretion. Neurons depend on microtubules for axoplasmic transport and when disrupted by vincristine, axonal degeneration and demyelination results. This occurs in a Wallerian-like and dying-back fashion and is reversible, unless nerve degeneration reaches the perikaryon [1]. The most common clinical consequences of neurotoxicity include loss of deep tendon reflexes, which may be associated with foot drop and gait disturbance, jaw pain, constipation and ileus. Non-neural side effects are markedly less common but can include alopecia and myelosuppression [1]. The lack of myelosuppression at standard doses reflects the inability to dose escalate due to neurotoxicity. The high level of cytotoxicity is notable despite low doses, unlike most other agents where myelosuppression is the dose limiting toxicity. On the basis that in general, paediatric cancer regimens are most effective when high dose intensity is achieved it seems probable that the effective- ness of vincristine could be enhanced if neurotoxicity was reduced or at least doses maintained. Moreover, dose individualisation could lead to increased activity in some or reduced toxicity in others, thus enhancing the therapeutic index. Despite its widespread clinical use, much remains unknown about such an important cytotoxic drug. Gidding et al. [1] published an excellent and extensive review of vincristine in 1999 but in the past decade, a number of studies have enhanced the understanding of vincristine’s pharmacokinetics, pharmacodynamics and pharma- cogenetics and could provide a basis for dose individualisation. New clinical applications for vincristine and strategies to prevent vincristine-related neurotoxicity have also been explored in recent years. CLINICAL USES Adult and Paediatric Cancer The role of vincristine in multi-modality chemotherapy regimens remains almost unparalleled in both adult and paediatric oncology practice. As with many drugs however, the precise role of vincristine in combination chemotherapy has not been demon- strated in randomised trials. There has, however, been debate as to the benefit of vincristine and steroid pulses in maintenance therapy for childhood acute lymphoblastic leukaemia (ALL), with a major study by Conter et al. [3] showing that children with intermediate risk ALL treated with intensive BFM chemotherapy did not benefit from the addition of maintenance pulses of vincristine and dexamethasone. Use in Non-Malignant Disease Vincristine has emerged as an effective agent in the treatment of haemangiomas, (including haemangioendothelioma and Kaposi- form haemangioendothelioma), particularly those complicated by the Kasabach – Merritt syndrome (KMS) of thrombocytopaenia and consumptive coagulopathy. The high tubulin content of endothelial cells, together with the proliferating nature of haemangiomas makes vincristine a logical agent to use for these lesions [2]. Since the first two reported cases successfully treated with weekly vincristine in 1995, there have been at least 22 case reports of successful vincristine therapy for haemangioma and KMS described in the literature [2,4–24]. The most frequent dose of vincristine described is 0.05 mg/kg/ dose or 1–1.5 mg/m 2 /dose given weekly initially with variable tapering regimens. One of the largest retrospective international studies involved 15 patients with haemangiomas and KMS refractory to prior treatments. Vincristine therapy (1–1.5 mg/m 2 or 0.05–0.065 mg/kg weekly with variable tapers) improved thrombocytopaenia and fibrinogen levels in all patients and significantly reduced the size of the lesions in 13 patients (87%) [8]. All four patients (27%) who relapsed were successfully Vincristine is one of the most widely used and more effective drugs in paediatric oncology. The dose-limiting toxicity of neuro- pathy, lack of proven neuroprotective measures and an incomplete understanding of the pharmacokinetics and pharmacogenetics of vincristine have limited its therapeutic potential. Recent advances in the understanding of vincristine pharmacokinetics and pharmaco- genetics, and potential methods of preventing neurotoxicity are reviewed which could enable dose escalation and dose individual- isation in order to enhance the therapeutic index. Pediatr Blood Cancer 2009;53:1180–1187. ß 2009 Wiley-Liss, Inc. Key words: cancer pharmacology; chemotherapy neurotoxicities; pharmacogenetics; pharmacokinetics; vincristine ß 2009 Wiley-Liss, Inc. DOI 10.1002/pbc.22161 Published online 8 July 2009 in Wiley InterScience (www.interscience.wiley.com) —————— 1 Section of Paediatric Oncology, The Institute of Cancer Research, Sutton, United Kingdom; 2 Queensland Children’s Cancer Centre, Royal Children’s Hospital, Brisbane, Queensland, Australia *Correspondence to: Andrew Moore, Clinical Research Fellow, Section of Paediatric Oncology, The Institute of Cancer Research, 15 Cotswold Rd, Sutton, Surrey SM2 5NG, United Kingdom. E-mail: [email protected] Received 16 February 2009; Accepted 26 May 2009

Vincristine: Can its therapeutic index be enhanced?

Embed Size (px)

Citation preview

Page 1: Vincristine: Can its therapeutic index be enhanced?

Pediatr Blood Cancer 2009;53:1180–1187

REVIEWVincristine: Can Its Therapeutic Index Be Enhanced?

Andrew Moore, MBBS1* and Ross Pinkerton, MD, FRCPCH

2

INTRODUCTION

Although first used successfully in adults and children as a

cytotoxic agent in 1962, vincristine has been known for its

medicinal properties since the 17th century. Extracts from the plant

Vinca rosea have been described in medicinal folklore as being

effective for the treatment of haemorrhage, scurvy, toothache,

wound healing, diabetic ulcers and hyperglycaemia [1]. Vincristine

exerts its cytotoxic effects via interference with microtubule

formation and mitotic spindle dynamics, disruption of intracellular

transport and decreased tumour blood flow, with the latter probably

as a consequence of anti-angiogenesis [1,2]. The ultimate result of

vincristine-mediated cellular disruption is apoptosis [1].

The most frequent and clinically important side-effect of

vincristine is neurotoxicity, characteristically resulting in an

autonomic and peripheral sensory-motor polyneuropathy. Central

nervous system toxicity is uncommon and can include the syndrome

of inappropriate anti-diuretic hormone (SIADH) secretion. Neurons

depend on microtubules for axoplasmic transport and when

disrupted by vincristine, axonal degeneration and demyelination

results. This occurs in a Wallerian-like and dying-back fashion and

is reversible, unless nerve degeneration reaches the perikaryon [1].

The most common clinical consequences of neurotoxicity include

loss of deep tendon reflexes, which may be associated with foot drop

and gait disturbance, jaw pain, constipation and ileus. Non-neural

side effects are markedly less common but can include alopecia and

myelosuppression [1].

The lack of myelosuppression at standard doses reflects the

inability to dose escalate due to neurotoxicity. The high level of

cytotoxicity is notable despite low doses, unlike most other agents

where myelosuppression is the dose limiting toxicity. On the basis

that in general, paediatric cancer regimens are most effective when

high dose intensity is achieved it seems probable that the effective-

ness of vincristine could be enhanced if neurotoxicity was reduced

or at least doses maintained. Moreover, dose individualisation could

lead to increased activity in some or reduced toxicity in others, thus

enhancing the therapeutic index.

Despite its widespread clinical use, much remains unknown

about such an important cytotoxic drug. Gidding et al. [1] published

an excellent and extensive review of vincristine in 1999 but in the

past decade, a number of studies have enhanced the understanding

of vincristine’s pharmacokinetics, pharmacodynamics and pharma-

cogenetics and could provide a basis for dose individualisation.

New clinical applications for vincristine and strategies to prevent

vincristine-related neurotoxicity have also been explored in recent

years.

CLINICAL USES

Adult and Paediatric Cancer

The role of vincristine in multi-modality chemotherapy

regimens remains almost unparalleled in both adult and paediatric

oncology practice. As with many drugs however, the precise role of

vincristine in combination chemotherapy has not been demon-

strated in randomised trials. There has, however, been debate as to

the benefit of vincristine and steroid pulses in maintenance therapy

for childhood acute lymphoblastic leukaemia (ALL), with a major

study by Conter et al. [3] showing that children with intermediate

risk ALL treated with intensive BFM chemotherapy did not benefit

from the addition of maintenance pulses of vincristine and

dexamethasone.

Use in Non-Malignant Disease

Vincristine has emerged as an effective agent in the treatment of

haemangiomas, (including haemangioendothelioma and Kaposi-

form haemangioendothelioma), particularly those complicated by

the Kasabach–Merritt syndrome (KMS) of thrombocytopaenia and

consumptive coagulopathy. The high tubulin content of endothelial

cells, together with the proliferating nature of haemangiomas makes

vincristine a logical agent to use for these lesions [2]. Since the first

two reported cases successfully treated with weekly vincristine

in 1995, there have been at least 22 case reports of successful

vincristine therapy for haemangioma and KMS described in the

literature [2,4–24].

The most frequent dose of vincristine described is 0.05 mg/kg/

dose or 1–1.5 mg/m2/dose given weekly initially with variable

tapering regimens. One of the largest retrospective international

studies involved 15 patients with haemangiomas and KMS

refractory to prior treatments. Vincristine therapy (1–1.5 mg/m2

or 0.05–0.065 mg/kg weekly with variable tapers) improved

thrombocytopaenia and fibrinogen levels in all patients and

significantly reduced the size of the lesions in 13 patients (87%)

[8]. All four patients (27%) who relapsed were successfully

Vincristine is one of the most widely used and more effectivedrugs in paediatric oncology. The dose-limiting toxicity of neuro-pathy, lack of proven neuroprotective measures and an incompleteunderstanding of the pharmacokinetics and pharmacogenetics ofvincristine have limited its therapeutic potential. Recent advances in

the understanding of vincristine pharmacokinetics and pharmaco-genetics, and potential methods of preventing neurotoxicity arereviewed which could enable dose escalation and dose individual-isation in order to enhance the therapeutic index. Pediatr BloodCancer 2009;53:1180–1187. � 2009 Wiley-Liss, Inc.

Key words: cancer pharmacology; chemotherapy neurotoxicities; pharmacogenetics; pharmacokinetics; vincristine

� 2009 Wiley-Liss, Inc.DOI 10.1002/pbc.22161Published online 8 July 2009 in Wiley InterScience(www.interscience.wiley.com)

——————1Section of Paediatric Oncology, The Institute of Cancer Research,

Sutton, United Kingdom; 2Queensland Children’s Cancer Centre,

Royal Children’s Hospital, Brisbane, Queensland, Australia

*Correspondence to: Andrew Moore, Clinical Research Fellow,

Section of Paediatric Oncology, The Institute of Cancer Research, 15

Cotswold Rd, Sutton, Surrey SM2 5NG, United Kingdom.

E-mail: [email protected]

Received 16 February 2009; Accepted 26 May 2009

Page 2: Vincristine: Can its therapeutic index be enhanced?

retreated with additional vincristine [8]. Gowans et al. [25] have also

described the case an infant with a potentially malignant

haemangiopericytoma successfully treated with vincristine

(0.05 mg/kg), doxorubicin, actinomycin-D and cyclophosphamide.

PHARMACOKINETICS

Despite a number of pharmacokinetic studies over the past

decade, the dosing strategy of vincristine, including the dose-

capping at 2 mg, largely remains empirical. Pharmacokinetic

research during the past 10 years has involved both the paediatric

and adolescent population. In a study of 32 children with a median

age of 4.6 years (range 0–16) receiving weekly vincristine for ALL,

non-Hodgkin lymphoma (NHL) or Wilms tumour, there was wide

inter- and intra-patient variability in distribution and elimination

half-life (t1/2), clearance, volume of distribution at steady state

and area under the concentration-time curve (AUC). The median

clearance of vincristine based on all 169 doses in the 32 children was

333 ml/min/m2 (range 34–830). Although vincristine clearance in

infants less than 1 year of age was lower compared to older children,

the study could not demonstrate any significant influence of age

above 1 year on vincristine clearance. On multivariate analysis,

diagnosis was the only statistically significant factor affecting

vincristine clearance with ALL/NHL patients receiving concom-

itant steroids having faster clearance rates compared to patients with

Wilms tumour (mean, 381� 140 ml/min/m2 vs. 258� 120 ml/min/

m2; P¼ 0.0092) [26].

A Nordic study of 98 children with newly diagnosed ALL also

failed to demonstrate any correlation between vincristine pharma-

cokinetics and age, sex, body surface area (BSA), body mass index,

dosage per m2 or biochemical variables [27]. Children were found to

reach a BSA of 1 m2 at 8–9 years of age and since the dose in the

protocol was 2.0 mg/m2 with a maximum dose of 2.0 mg, several

teenage patients only received approximately 1.0 mg/m2. As a

consequence, the vincristine AUC negatively correlated with age,

but when corrected for BSA, this correlation was lost. Likewise,

there was no difference in median total body clearance for patients

older (n¼ 26) or younger (n¼ 72) than 10 years of age (371 versus

353 ml/min/m2, respectively) [27]. Six patients with Down

syndrome were included in the study, although no pharmacokinetic

differences were detected between Down syndrome and non-Down

syndrome patients [28]. Long term follow-up (median 10.5 years,

range 7.3–12) of this patient cohort demonstrated that patients with

standard risk precursor B cell ALL had a significantly higher relative

risk (RR) of relapse if their vincristine clearance was above the

median (RR 5.2; P¼ 0.036) or AUC below the median (RR 5.8;

P¼ 0.025) [29]. Although there was a similar trend overall, no

significant difference in relapse risk was detected for patients with

intermediate or high risk precursor B cell ALL. The authors suggest

the significant relationship between relapse risk and vincristine

pharmacokinetics for standard risk patients is likely due to the

homogenous nature of this patient group and the increased relative

importance of vincristine in standard risk protocols compared to

intermediate and high risk protocols that utilise more intensive

multi-agent chemotherapy strategies [29].

Studies assessing high doses of vincristine are limited. In 1994,

Haim et al. [30] reported the results of a trial administering full dose

vincristine (1.4 mg/m2 with no limit to the first dose) to 104 adults

with Hodgkin disease and NHL (median age 52 years, range 18–72)

and acceptable performance status (median WHO score 1, range

0–3). Patients received multi-agent chemotherapy with vincristine,

vinblastine, prednisone, methotrexate, calcium leucovorin, doxor-

ubicin, cyclophosphamide, etoposide, mechlorethamine, procarba-

zine and bleomycin, usually as part of ProMACE/MOPP, CHOP and

MOPP/ABV regimens. Overall, the average dose of vincristine was

2.55 mg for men and 2.29 mg for women, with 90% of patients

receiving a first dose >2 mg, but only 7/23 (30%) receiving >2 mg

with the eighth course of vincristine, due to toxicity-related dose

reductions [30]. Symptoms of vincristine neuropathy in this adult

cohort were almost universal; 92% reported at least one symptom,

with 78% experiencing paraesthesia/dysaesthesia (13% grade 2–3),

59% constipation (10% grade 3–4), 45% muscle cramps, 38%

motor weakness (13% grade 3, none grade 4), 24% jaw pain, 23%

muscle/bone pain, 15% of males erectile/ejaculatory dysfunction,

14% postural hypotension and 9% taste disturbance [30]. Despite

the frequency of symptoms, the authors reported vincristine

neuropathy as being mostly reversible, with significant symptom

improvement occurring within weeks of vincristine cessation or

dose reduction and concluded that full dose vincristine was still

feasible in patients older than 65 years [30].

Only one study reporting the use of high-dose vincristine has

been published in the past decade. Kellie et al. adopted a schedule

used in high dose therapy prior to autologous marrow rescue

[31], administering cyclophosphamide (65 mg/kg) on day 1, then

vincristine as a 1.5 mg/m2 bolus on day 2, followed immediately by

a continuous vincristine infusion of 0.5 mg/m2 per 24 hr for 96 hr to

16 children (median age 4.8 years, range 1.7–15.8 years) with

central nervous system tumours [32]. Fifteen patients received two

courses of treatment at 21–28 day intervals. Such a continuous

infusion of vincristine achieved a steady state after 1 hr and the mean

total-body clearance following the infusion of 203 ml/m2/min was

similar to an earlier study of vincristine monotherapy in leukaemia

patients [32,33]. The total dose of 3.5 mg/m2 delivered over 4 days

was reported as being well tolerated, with 61% of treatment courses

resulting in grade 1 or 2 neurotoxicity and only 1 patient (6%)

experiencing grade 3 neurotoxicity. A single dose of cyclo-

phosphamide (65 mg/kg) was administered on the day prior to each

vincristine infusion and, not surprisingly, the majority of patients

experienced neutropaenia (75% �0.5� 109/L; 63% �0.5� 109/L)

[32]. The authors highlight the potential therapeutic benefit of

having vincristine present when cell replication is occurring within

tumours composed of heterogenous cell populations [32]. Whether

this justifies the expense and resource demands of prolonged

vincristine infusions requires further evaluation.

The cytochrome P450 3A4 (CYP 3A4) enzyme system is heavily

involved in the metabolism of vincristine and is induced by

corticosteroids [34–36]. The induction of CYP3A4 by steroids

likely accounts for the faster clearance rates in patients receiving

concomitant steroids compared to patients who do not receive high

dose steroids as part of multi-agent chemotherapy as demonstrated

by Gidding et al. [26]. In a window study of vincristine monotherapy

in 70 children over 1 year of age with untreated ALL, Groninger

et al. [33] demonstrated a median vincristine clearance of 228 ml/

min/m2 (interquartile range 128–360) which is markedly slower

than rates reported by studies where patients received concomitant

steroids for ALL. Interestingly, vincristine clearance was faster in

patients whose leukaemic blasts had >50 chromosomes compared

to patients with diploid or hyperdiploid (46–50 chromosomes)

leukaemia [33]. Further analysis of this patient group however,

failed to detect any correlation between the antileukaemic effect of

Pediatr Blood Cancer DOI 10.1002/pbc

Vincristine: Can Its Therapeutic Index Be Enhanced? 1181

Page 3: Vincristine: Can its therapeutic index be enhanced?

vincristine and vincristine clearance, elimination half-life or peak

plasma concentration [37].

Published studies have in general utilised high performance

liquid chromatography (HPLC) assays. The use of mass spectrom-

etry which permits the clear separation of parent drug from

metabolite should provide more accurate information about

pharmacokinetics and in particular shed more light on the relevance

of metabolites to toxicity and efficacy.

PHARMACOGENETICS

A number of in vitro studies have highlighted the complex

pharmacogenetics involved in vincristine metabolism. Vincristine is

predominantly metabolised to one metabolite, M1, in the liver by the

cytochrome P450 (CYP) enzymes CYP3A4 and CYP3A5. CYP3A5

is up to 14 times more efficient than CYP3A4 and has a number

of common genetic polymorphisms, including CYP3A5*1,

CYP3A5*3, CYP3A5*6 and CYP3A5*7 [38–41]. CYP3A5 produc-

tion is heavily dependent on the presence of at least one functioning

CYP3A5*1 allele, without this very little CYP3A5 is produced and

vincristine metabolism will be largely dependent on CYP3A4.

Conversely, the presence of one or two active CYP3A5*1 alleles

results in high expression of CYP3A5, accounting for approxi-

mately 40% of all CYP3A protein activity and significant vincristine

metabolism [41,42]. The CYP3A5*3 allele is most common in

Caucasians (allele frequency 89–94%) and encodes abnormal

splicing resulting in an absence of CYP3A5 enzyme [34].

The expression of CYP3A5 varies considerably according to

race, with 75% of African-Americans being high expressors,

compared to 47% of East Asians and 19% of Caucasians [38].

African-American children have been shown to have inferior

survival compared to Caucasian children with ALL [43,44]. It is

possible the high expression of CYP3A5 in African-Americans and

East Asians results in increased clearance of vincristine and may

contribute to this disparity in survival. In light of the finding that

African-American children with intermediate risk ALL treated on

the CCG-1891 protocol had a markedly inferior event-free survival

(EFS) at 6 years compared to Caucasian children (54% vs. 82%,

respectively) [43], Aplenc et al. [45] performed a retrospective

analysis of relapse and CYP3A genotype. CYP3A variants were not

found to be associated with an increased risk of relapse and,

inexplicably patients with CYP3A5*3 genotypes had a reduced risk

of peripheral neuropathy [45]. This result is counter-intuitive if

neurotoxicity is related to PK of parent drug since as there is no

CYP3A5 enzyme activity with CYP3A5*3 vincristine metabolism

would be expected to be less rapid.

Evidence for a racial difference in vincristine pharmacokinetics

was demonstrated by Renbarger et al. [46] in a retrospective study of

92 non-Hispanic Caucasian and 21 African-American children with

ALL. When compared to Caucasian children, African-American

children had lower average neurotoxicity grades (1 vs. 2.72, P¼< 0.0001), less vincristine-related neurotoxicity (4.8% vs. 34.8%,

P¼ 0.007), fewer vincristine doses reduced due to vincristine-

related neurotoxicity (0.1% vs. 4.0%, P¼ < 0.0001) and fewer

vincristine doses omitted due to vincristine-related neurotoxicity

(0.1% vs. 1.2%, P¼ 0.01). Patients were treated on various

protocols and although the authors report there was no significant

difference in cumulative vincristine dose, there was a trend toward

lower doses in African Americans (42.44� 11.62 mg/m2 vs.

48.52� 14.25 mg/m2; P¼ 0.07) and it was not stated whether

individual vincristine doses were capped. Although the African-

American children were significantly older than the Caucasian

children (mean age 8.2� 4.80 years vs. 4.95� 3.08; P¼ 0.0002),

the authors acknowledged this difference and did not believe it

impacted on the overall findings. As discussed above, a body surface

area of 1.0 m2 is reached at approximately 8–9 years of age [27] so

it is possible that the impact of relative under-dosing in these

older African-American children may have been underestimated.

Furthermore, race was used as a retrospective surrogate marker of

CYP3A5 genotype. Despite these potential confounders however,

the difference in vincristine related toxicity in the Renbarger et al.

study is compelling.

The syndrome of inappropriate anti-diuretic syndrome (SIADH)

resulting in hyponatraemia is a rare, but well recognised side effect

of vincristine [47]. A retrospective review of a pharmaceutical

company’s (Eli Lilly and Company, Indianapolis, IN) global safety

database revealed a marked over-representation of Asian patients

with SIADH and/or hyponatraemia (90% Asian, 8% Caucasian,

2% Black). Major limitations of this study include its retrospective

analysis of a spontaneously reported adverse event register and the

finding that two thirds of the cases were reported from Japan.

However, when the cases of SIADH reported from the United States

(US) were analysed, 75% of cases where race was reported involved

Asian patients. If all the US cases where race was unknown were

assumed to be non-Asian, Asian patients would still have accounted

for 41% of SIADH cases reported from the US [47]. A biological

explanation for the apparent predisposition of Asian patients to

SIADH was not investigated and further research is warranted.

The multi-drug resistance MDR1 gene (also called ATP-binding

cassette subfamily B, ABCB1) encodes for Permeability-glycopro-

tein (P-glycoprotein), a transmembrane efflux pump transporting

drugs from the intracellular to the extracellular compartment and

implicated in the resistance and pharmacokinetics of many drugs,

including vincristine [48,49]. P-glycoprotein is found on a variety of

cells including hepatocytes, renal tubular cells, enterocytes, various

blood cells including haematopoietic stem cells and cells lining

blood-tissue barriers including the brain, cerebrospinal fluid, testes,

ovaries and placenta (reviewed in Ref. [48]). Preclinical models

using P-glycoprotein inducers and inhibitors have demonstrated a

role for P-glycoprotein in the biliary excretion and renal clearance of

vincristine [50–52].

A number of functional single nucleotide polymorphisms

(SNPs) have been identified for MDR1, raising the possibility that

genetic variations in this gene may contribute to inter-patient

variation in vincristine pharmacokinetics, treatment response and

toxicity [48,49]. Few clinical studies have tested these hypotheses

however. When SNPs of the MDR1 gene were retrospectively

analysed for 52 children with ALL treated with vincristine

monotherapy, no association was found between the SNPs

C3435T or G2677T and vincristine pharmacokinetics or toxicity

[49]. Furthermore, when assessed prospectively, MDR1 gene

expression in childhood ALL blasts at diagnosis did not correlate

with levels of minimal residual disease at the end of induction [53].

STATEGIES TO REDUCE TOXICITY

Liposomal Vincristine

Liposomal formulations of drugs, ranging from cosmetics to

cytotoxics and antifungal agents have been used in attempt to

optimise drug delivery to target tissues and minimise toxicity, with

Pediatr Blood Cancer DOI 10.1002/pbc

1182 Moore and Pinkerton

Page 4: Vincristine: Can its therapeutic index be enhanced?

liposomal doxorubicin and daunorubicin being the most extensively

studied anti-cancer drugs [54]. Vincristine has been encapsulated

in both sphingomyelin/cholesterol liposomes (sphingosomal vin-

cristine) and the more permeable distearoylphosphatidylcholine/

cholesterol (DSPC/Chol) liposomes [54]. An early phase I trial

of sphingosomal vincristine reported a maximum tolerated dose

(MTD) of 2.4 mg/m2 and recommended phase II dose of 2.0 mg/m2,

with pain and constipation being the dose-limiting toxicities [55].

Boehlke and Winter [54] reviewed the use of sphingosomal

vincristine in seven trials involving patients with solid tumours,

ALL, Hodgkin disease and NHL. They concluded that sphingoso-

mal vincristine had a favourable side effect profile, responses were

demonstrated in heavily pre-treated patients and excellent outcomes

were seen in studies that used sphingosomal vincristine as part of

CHOP or R-CHOP multi-agent therapy for NHL [54].

Bedikian et al. [56,57] have subsequently reported the use of

vincristine sulphate liposome infusion (VSLI) in 27 patients with

metastatic melanoma. At doses to provide 2.0 mg/m2 vincristine

without an upper dose cap, VSLI was reportedly well tolerated,

although approximately 65% of patients developed VSLI-associ-

ated fever within 48 hr of infusion [57]. Other frequent toxicities

included constipation in 48% of patients (grade 3 in 15%), muscle

cramps in 30%, anxiety/psychiatric disorders in 30% (grade 3 in

7%) and paraesthesia in 22% (grade 3 in 4%). Grade 3 arthralgia and

infection occurred in 1 patient each (4%) but no grade 4 toxicities

were reported. Grade 1–2 neutropaenia was the most common

haematological toxicity although its frequency was not quantified

[57]. Disease control was achieved in 31% of patients with

a median time to progression of 1.9 months (95% CI: 1.8–

2.2 months). One patient with two isolated pulmonary metastases,

arising from a previously treated and enucleated uveal primary,

achieved a complete response after 9 months of therapy (VSLI given

every 2 weeks). At the time of Bedikian et al.’s [57] report,

almost 3 years after completing 24 cycles of VSLI, he remained

disease-free.

Currently there are three active (two recruiting) phase II trials

registered on ClinicalTrials.gov, investigating the use of VSLI

in relapsed ALL (NCT00495079), metastatic malignant uveal

melanoma (NCT00506142) and relapsed/refractory NHL (NCT

00006383) [58].

Charcot-Marie-Tooth Disease

The association between severe vincristine-related neurotoxicity

and underlying neuropathies such as Charcot-Marie-Tooth disease

(CMT) is well established [1]. Numerous case reports in the past

decade have highlighted the potential for previously undiagnosed

CMT (including type 2 CMT) being unmasked by vincristine

[59–67]. Despite the strong association with type 1A CMT,

Ajitsaria et al. [68] recently reported the successful administration

of vincristine to a 5-year-old boy with Wilms tumour and previously

diagnosed x-linked type 1 CMT. The complex relationship between

neuropathy and vincristine pharmacology is highlighted in Porter

et al.’s [69] report of vincristine-induced neuropathy in a child with

previously undiagnosed x-linked type 1 CMT receiving concom-

itant voriconazole. Although the unmasking of CMT with vincris-

tine often occurs in the absence of a family history, the need for

taking a thorough family history prior to vincristine therapy remains

paramount.

Important Drug Interactions

Drugs know to potentiate vincristine-induced neurotoxicity

through inhibition of CYP3A enzymes or inhibition of P-glyco-

protein, include nifedipine, cyclosporine, itraconazole, voricona-

zole and possibly posaconazole and CYP3A4-inducing highly

active antiretroviral therapy (HAART) [69–80]. Important vincris-

tine drug interactions are summarised in Table I. The list of drugs

in Table I is by no means exhaustive. Physician and pharmacist

vigilance for potential drug interactions with vincristine and

appropriate dose reduction or the avoidance of concomitant dosing

is of obvious importance.Despite numerous case reports of

increased neurotoxicity in the setting of concomitant azole and

vincristine therapies, there are currently no evidence-based guide-

lines addressing this potential drug interaction, nor the need for

vincristine dose adjustment following an episode of severe neuro-

toxicity. Specific recommendations are therefore based upon expert

opinion and available case reports. Moreover, withholding or

reducing doses of vincristine may not result in a rapid improvement

of neurotoxicity symptoms such as foot drop, and it is important to

not risk effectiveness for what are often reversible symptoms [30].

For patients receiving itraconazole, voriconazole or posaconazole it

may be prudent to withhold these agents from the day prior until

the day after the planned vincristine dose if the antifungal agent is

being used for prophylaxis or empiric therapy. Alternatively, a

substitute antifungal agent from another class may be administered.

For patients with proven, probable or possible invasive fungal

infection where azole therapy is required and alternative antifungal

therapies are contraindicated or not appropriate, dose-reduction of

vincristine should be considered on an individual basis [81].

Pediatr Blood Cancer DOI 10.1002/pbc

TABLE I. Clinically Important Drug Interactions With Vincristine

Drug

Effect on vincristine

concentration Mechanism of interaction References

Aprepitant Variable CYP3A4 Inhibition then Induction [82]

Azole antifungals Increase CYP3A inhibition [69-79,81]

Nifedipine Increase CYP3A and P-glycoprotein inhibition [70,71]

Cyclosporin A Increase CYP3A and P-glycoprotein inhibition [70]

Erythromycin Increase CYP3A inhibition [70]

HAART Variable CYP3A inhibition and induction [80]

Corticosteroids Decrease CYP3A induction [34-36]

Carbamazepine Decrease CYP3A4 induction [83]

Phenytoin Decrease CYP3A induction [83]

CYP, cytochrome P450; HAART, highly active antiretroviral therapy; VCR, vincristine.

Vincristine: Can Its Therapeutic Index Be Enhanced? 1183

Page 5: Vincristine: Can its therapeutic index be enhanced?

Neuroprotective Agents

Gidding et al. [1] reviewed the use of neuroprotective agents for

vincristine-related neurotoxicity including folinic acid, vitamin B1,

B6 and B12, isaxonine, glutamic acid, gangliosides, adrenocortico-

tropin hormone (ACTH) analogues, insulin-like growth factor 1

(IGF-1) and nerve growth factor (NGF) and concluded there was

insufficient evidence to recommend the use of any of these agents

outside the experimental setting. Over the past decade, research in

this area has continued with preclinical investigation of the effect of

agents such as FK506, verapamil, amiloride, pralidoxime, brady-

kinin receptor antagonists, mexiletine and propentofylline [84–88].

There has also been interest in understanding the protective effect of

the gene for slow Wallerian degeneration (Wlds) which could have

clinical implications [89–91].

There have been few recent major clinical trials of neuro-

protective agents. Koeppen et al. [92] investigated the effect of the

ACTH (4–9) analogue Org 2766 on the neuropathy-free interval in

150 patients being treated with vincristine as part of multi-agent

chemotherapy for NHL or Hodgkin disease. Adult patients (mean

age 46.6 years) expected to receive a cumulative vincristine dose of

at least 8 mg were randomised to receive placebo or Org 2766 before

and after each vincristine dose and 3 to 4 weeks after cessation

of vincristine therapy. No significant differences were observed

between the placebo and treatment groups and for the 80 patients

treated uniformly with CEBOPP chemotherapy, Org 2766 had no

influence on tumour response [92].

Three NIH-registered clinical trials aimed at minimising

vincristine neuropathy are currently open and recruiting patients

[58]. All three trials are recruiting patients with both solid and

haematological cancers. A randomised, placebo-controlled, single-

centre phase II trial (NCT00365768) of glutamine in children and

adolescents (5–21 years, estimated enrolment 100 patients) with

established grade I peripheral neuropathy, is administering oral

placebo or oral glutamine twice daily for 21 days following a dose of

vincristine [58]. A larger multi-centre, randomised phase III study

(NCT00369564, ACCL0731) of glutamic acid in children and

adolescents (3–20 years, estimated enrolment 208 patients) is

administering oral placebo or oral glutamic acid three times daily

from before the first dose of vincristine until either 5 or 10 weeks

with the primary objective being to compare the reduction in

neurotoxicity as measured by a scored neurological examination.

For this study, patients are not permitted regular prophylactic

laxatives or itraconazole [58]. Finally, vitamin B6 and B12 are being

assessed in a randomised, phase III study of adults (NCT00659269,

age over 18 years, estimated enrolment 228 patients) being treated

with heavy metals, taxanes or vinca alkaloids. Patient stratification

for this study is based on treatment and the presence of pre-existing

neuropathy, however there is no true placebo arm, since the

experimental arm involves intramuscular injections of vitamin B12

(1 mg) plus oral pyridoxine (50 mg tid), whilst the active comparator

arm gives a multivitamin containing no more than 10 mg of

pyridoxine or 10 mg of vitamin B12 [58].

CONCLUSIONS

Vincristine continues to be an invaluable agent in the treatment

of cancer. The narrow therapeutic window and apparent dose-

limiting toxicity of neuropathy has limited its potential and led to

largely empiric dose capping at 2 mg. Such dose-restriction results

in many patients receiving a suboptimal dose and in adolescents, this

may contribute to poorer prognosis for diseases where vincristine

plays an important role in upfront therapy such as ALL and sarcoma.

Maximal doses of 2.5 mg have been successfully used in paediatric

ALL protocols without an increase in toxicity, although the

influence of steroids in ALL protocols may increase the toxicity

threshold of vincristine in this setting [27,93]. Suboptimal dosing of

chemotherapy has been known to produce inferior outcomes for

children with cancer since the 1960s [94]. Despite the results of

numerous pharmacokinetic studies of vincristine over the past

decade, clinical trials of multi-agent chemotherapy have not

addressed vincristine dosing. This is likely due to patient numbers

limiting the number of possible hypotheses to test, as well as a focus

on other components of chemotherapy, such as anthracycline dose

reduction and asparaginase dose optimisation. Since there is a real

potential for vincristine under-dosing, especially in adolescents,

future clinical trials should specifically address vincristine dosing.

Research on the pharmacokinetics and pharmacogenetics

of vincristine is ongoing. The authors have recently completed a

study examining the relationship between vincristine pharmaco-

kinetics, CYP3A genotype and toxicity. The Children’s Oncology

Group is currently undertaking a similar study of vincristine and

actinomycin D (NCT00674193, ADVL06B1) [58]. Although

pharmacogenetics offers the hope of providing ‘personalised

chemotherapy’, the challenge remains to incorporate pharmacoge-

netically guided dosing for individual drugs in the context of multi-

agent clinical trials such as those for ALL in children and

demonstrate an improvement in survival [95]. Furthermore, the

results of multi-agent trials with pharmacogenetically guided

dosing for an individual drug may be protocol specific and not

broadly applicable [95]. More reasonable objectives for favourable-

risk diseases might be to maintain survival but with reduced

vincristine-related toxicity. The results of current trials investigating

neuroprotective agents such as glutamic acid may increase the

therapeutic window of vincristine. Taking into account age, race,

CYP genotype and utilising effective neuroprotectants the aim is

ultimately to anticipate toxicity and pre-emptively modify dose

accordingly or enable dose escalation to maintain or maximise

effectiveness.

REFERENCES

1. Gidding CE, Kellie SJ, Kamps WA, et al. Vincristine revisited. Crit

Rev Oncol Hematol 29:267–287.

2. Perez J, Pardo J, Gomez C. Vincristine: An effective treatment of

corticoid-resistant life-threatening infantile hemangiomas. Acta

Oncol 2002;41:197–199.

3. Conter V, Valsecchi MG, Silvestri D, et al. Pulses of vincristine and

dexamethasone in addition to intensive chemotherapy for children

with intermediate-risk acute lymphoblastic leukaemia: A multi-

centre randomised trial. Lancet 2007;369:123–131.

4. Vin-Christian K, McCalmont TH, Frieden IJ. Kaposiform

hemangioendothelioma. An aggressive, locally invasive vascular

tumor that can mimic hemangioma of infancy. Arch Dermatol

1997;133:1573–1578.

5. Yang YH, Lee PI, Lin KH, et al. Absolute ethanol embolotherapy

for hemangioma with Kasabach-Merritt syndrome. Zhonghua

Minguo xiao er ke yi xue hui za zhi 1998;39:51–54.

6. Enjolras O, Wassef M, Dosquet C, et al. Kasabach-Merritt syndrome

on a congenital tufted angioma. Ann Dermatol Venereol 1998;125:

257–260.

7. Hu B, Lachman R, Phillips J, et al. Kasabach-Merritt syndrome-

associated kaposiform hemangioendothelioma successfully treated

Pediatr Blood Cancer DOI 10.1002/pbc

1184 Moore and Pinkerton

Page 6: Vincristine: Can its therapeutic index be enhanced?

with cyclophosphamide, vincristine, and actinomycin D. Am J

Pediatr Hematol Oncol: Off J Am Soc Pediatr Hematol Oncol

1998;20:567–569.

8. Haisley-Royster C, Enjolras O, Frieden IJ, et al. Kasabach-merritt

phenomenon: A retrospective study of treatment with vincristine.

Am J Pediatr Hematol Oncol 2002;24:459–462.

9. Leal N, Lopez Santamaria M, Gamez M, et al. The multifocal

hepatic hemangioendothelioma. Is always a benign tumor? Cirugia

Pediatrica: Organo Oficial Sociedad Espanola Cirugia Pediatrica

2004;17:8–11.

10. Fawcett SL, Grant I, Hall PN, et al. Vincristine as a treatment for a

large haemangioma threatening vital functions. Br J Plast Surg

2004;57:168–171.

11. Lopez Gutierrez JC, Patron Romero M. Thoracic kaposiform

hemangioendothelioma. Four consecutive cases with distinct

outcome. Anal Pediatr 2005;63:72–76.

12. Hauer J, Graubner U, Konstantopoulos N, et al. Effective treatment

of kaposiform hemangioendotheliomas associated with Kasabach-

Merritt phenomenon using four-drug regimen. Pediatric Blood

Cancer 2007;49:852–854.

13. Taki M, Ohi C, Yamashita A, et al. Successful treatment with

vincristine of an infant with intractable Kasabach-Merritt syn-

drome. Pediatr Int 2006;48:82–84.

14. Yeung J, Somers G, Viero S, et al. Multifocal lymphangioendo-

theliomatosis with thrombocytopenia. J Am Acad Dermatol 2006;

54:S214–S217.

15. Delesalle F, Staumont D, Houmany MA, et al. Pulse methyl-

prednisolone therapy for threatening periocular haemangiomas of

infancy. Acta Dermato-Venereologica 2006;86:429–432.

16. Thomson K, Pinnock R, Teague L, et al. Vincristine for the

treatment of Kasabach-Merritt syndrome: Recent New Zealand

case experience. NZ Med J 2007;120:U2418.

17. Herrero Hernandez A, Escobosa Sanchez O, Acha Garcia T.

Successful treatment with vincristine in PHACES syndrome. Clin

Transl Oncol: Off Publ Federation Spanish Oncol Soc Natil Cancer

Inst Mexico 2007;9:262–263.

18. Draper H, Diamond IR, Temple M, et al. Multimodal management

of endangering hepatic hemangioma: Impact on transplant

avoidance: A descriptive case series. J Pediatr Surg 2008;43:

120–125. discussion 126.

19. Yesudian PD, Parslew R, Klafowski J, et al. Tufted angioma-

associated Kasabach-Merritt syndrome treated with embolization

and vincristine. Plast Reconstr Surg 2008;121:692–693.

20. Abass K, Saad H, Kherala M, et al. Successful treatment of

Kasabach-Merritt syndrome with vincristine and surgery: A case

report and review of literature. Cases J 2008;1:9.

21. Walsh MA, Carcao M, Pope E, et al. Kaposiform hemangioendo-

thelioma presenting antenatally with a pericardial effusion. Am J

Pediatr Hematol Oncol: Off J Am Soc Pediatr Hematol Oncol

2008;30:761–763.

22. Enjolras O, Breviere GM, Roger G, et al. Vincristine treatment for

function- and life-threatening infantile hemangioma. Archives

Pediatrie: Organe Officiel Societe Francaise Pediatrie 2004;11:99–

107.

23. Shroff PK, Martin TW, Schmitz ML. Successful anesthetic

management of a child with an extensive facial hemangioma and

high output cardiac failure for placement of a central venous

catheter. Paediatr Anaesth 2006;16:77–81.

24. Perez Payarols J, Pardo Masferrer J, Gomez Bellvert C. Treatment

of life-threatening infantile hemangiomas with vincristine. N Engl

J Med 1995;333:69.

25. Gowans LK, Bentz ML, DeSantes KB, et al. Successful treatment

of an infant with constitutional chromosomal abnormality and

hemangiopericytoma with chemotherapy alone. Am J Pediatr

Hematol Oncol 2007;29:409–411.

26. Gidding CE, Meeuwsen-de Boer GJ, Koopmans P, et al. Vincristine

pharmacokinetics after repetitive dosing in children. Cancer

Chemother Pharmacol 1999;44:203–209.

27. Frost BM, Lonnerholm G, Koopmans P, et al. Vincristine in

childhood leukaemia: No pharmacokinetic rationale for dose

reduction in adolescents. Acta Paediatr 2003;92:551–557.

28. Lonnerholm G, Frost BM, Soderhall S, et al. Vincristine pharma-

cokinetics in children with Down syndrome. Pediatric Blood

Cancer 2009;52:123–125.

29. Lonnerholm G, Frost BM, Abrahamsson J, et al. Vincristine

pharmacokinetics is related to clinical outcome in children with

standard risk acute lymphoblastic leukemia. Br J Haematol 2008;

142:616–621.

30. Haim N, Epelbaum R, Ben-Shahar M, et al. Full dose vincristine

(without 2-mg dose limit) in the treatment of lymphomas. Cancer

1994;73:2515–2519.

31. Pinkerton CR, McDermott B, Philip T, et al. Continuous vincristine

infusion as part of a high dose chemoradiotherapy regimen: Drug

kinetics and toxicity. Cancer Chemother Pharmacol 1988;22:271–

274.

32. Kellie SJ, Koopmans P, Earl J, et al. Increasing the dosage of

vincristine: A clinical and pharmacokinetic study of continuous-

infusion vincristine in children with central nervous system tumors.

Cancer 2004;100:2637–2643.

33. Groninger E, Meeuwsen-de Boar T, Koopmans P, et al. Pharma-

cokinetics of vincristine monotherapy in childhood acute lympho-

blastic leukemia. Pediatr Res 2002;52:113–118.

34. van Schaik RH. CYP450 pharmacogenetics for personalizing

cancer therapy. Drug Resistance Updates 2008;11:77–98.

35. Pichard L, Fabre I, Daujat M, et al. Effect of corticosteroids on the

expression of cytochromes P450 and on cyclosporin A oxidase

activity in primary cultures of human hepatocytes. Mol Pharmacol

1992;41:1047–1055.

36. Guengerich FP. Cytochrome P-450 3 A4: Regulation and role in

drug metabolism. Annu Rev Pharmacol Toxicol 1999;39:1–17.

37. Groninger E, Meeuwsen-de Boer T, Koopmans P, et al. Vincristine

pharmacokinetics and response to vincristine monotherapy in an

up-front window study of the Dutch Childhood Leukaemia Study

Group (DCLSG). Eur J Cancer 2005;41:98–103.

38. Xie HG, Wood AJ, Kim RB, et al. Genetic variability in CYP3A5 and

its possible consequences. Pharmacogenomics 2004;5:243–272.

39. Dennison JB, Kulanthaivel P, Barbuch RJ, et al. Selective meta-

bolism of vincristine in vitro by CYP3A5. Drug Metab Dispos: Biol

Fate Chem 2006;34:1317–1327.

40. Dennison JB, Jones DR, Renbarger JL, et al. Effect of CYP3A5

expression on vincristine metabolism with human liver micro-

somes. J Pharmacol Exp Ther 2007;321:553–563.

41. Dennison JB, Mohutsky MA, Barbuch RJ, et al. Apparent high

CYP3A5 expression is required for significant metabolism of

vincristine by human cryopreserved hepatocytes. J Pharmacol Exp

Ther 2008;327:248–257.

42. Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A

promoters and characterization of the genetic basis of polymorphic

CYP3A5 expression. Nat Genet 2001;27:383–391.

43. Lange BJ, Bostrom BC, Cherlow JM, et al. Double-delayed

intensification improves event-free survival for children with

intermediate-risk acute lymphoblastic leukemia: A report from the

Children’s Cancer Group. Blood 2002;99:825–833.

44. Pollock BH, DeBaun MR, Camitta BM, et al. Racial differences

in the survival of childhood B-precursor acute lymphoblastic

leukemia: A Pediatric Oncology Group Study. J Clin Oncol 2000;

18:813–823.

45. Aplenc R, Glatfelter W, Han P, et al. CYP3A genotypes and

treatment response in paediatric acute lymphoblastic leukaemia.

Br J Haematol 2003;122:240–244.

Pediatr Blood Cancer DOI 10.1002/pbc

Vincristine: Can Its Therapeutic Index Be Enhanced? 1185

Page 7: Vincristine: Can its therapeutic index be enhanced?

46. Renbarger JL, McCammack KC, Rouse CE, et al. Effect of race on

vincristine-associated neurotoxicity in pediatric acute lympho-

blastic leukemia patients. Pediatric Blood Cancer 2008;50:769–

771.

47. Hammond IW, Ferguson JA, Kwong K, et al. Hyponatremia and

syndrome of inappropriate anti-diuretic hormone reported with the

use of Vincristine: An over-representation of Asians? Pharmacoe-

pidemiol Drug Safety 2002;11:229–234.

48. Marzolini C, Paus E, Buclin T, et al. Polymorphisms in human

MDR1 (P-glycoprotein): Recent advances and clinical relevance.

Clin Pharmacol Ther 2004;75:13–33.

49. Plasschaert SL, Groninger E, Boezen M, et al. Influence of

functional polymorphisms of the MDR1 gene on vincristine

pharmacokinetics in childhood acute lymphoblastic leukemia.

Clin Pharmacol Ther 2004;76:220–229.

50. Watanabe T, Miyauchi S, Sawada Y, et al. Kinetic analysis of

hepatobiliary transport of vincristine in perfused rat liver. Possible

roles of P-glycoprotein in biliary excretion of vincristine. J Hepatol

1992;16:77–88.

51. Watanabe T, Suzuki H, Sawada Y, et al. Induction of hepatic

P-glycoprotein enhances biliary excretion of vincristine in rats.

J Hepatol 1995;23:440–448.

52. Song S, Suzuki H, Kawai R, et al. Effect of PSC 833, a P-

glycoprotein modulator, on the disposition of vincristine and

digoxin in rats. Drug Metab Dispos: Biol Fate Chem 1999;27:689–

694.

53. Fedasenka UU, Shman TV, Savitski VP, et al. Expression of MDR1,

LRP, BCRP and Bcl-2 genes at diagnosis of childhood all:

Comparison with MRD status after induction therapy. Exp Oncol

2008;30:248–252.

54. Boehlke L, Winter JN. Sphingomyelin/cholesterol liposomal

vincristine: A new formulation for an old drug. Expert Opin Biol

Therapy 2006;6:409–415.

55. Gelmon KA, Tolcher A, Diab AR, et al. Phase I study of liposomal

vincristine. J Clin Oncol 1999;17:697–705.

56. Bedikian AY, Vardeleon A, Smith T, et al. Pharmacokinetics and

urinary excretion of vincristine sulfate liposomes injection in

metastatic melanoma patients. J Clin Pharmacol 2006;46:727–

737.

57. Bedikian AY, Papadopoulos NE, Kim KB, et al. A pilot study with

vincristine sulfate liposome infusion in patients with metastatic

melanoma. Melanoma Res 2008;18:400–404.

58. NIH. 2009 22/01/2009. ClinicalTrials.gov. www.clinicaltrials.gov.

Accessed 2009 22/01/2009.

59. Neumann Y, Toren A, Rechavi G, et al. Vincristine treatment

triggering the expression of asymptomatic Charcot-Marie-Tooth

disease. Med Pediatr Oncol 1996;26:280–283.

60. Nishikawa T, Kawakami K, Kumamoto T, et al. Severe neuro-

toxicities in a case of Charcot-Marie-Tooth disease type 2 caused

by vincristine for acute lymphoblastic leukemia. Am J Pediatr

Hematol Oncol 2008;30:519–521.

61. Schiavetti A, Frascarelli M, Uccini S, et al. Vincristine neuropathy:

Neurophysiological and genetic studies in a case of Wilms tumor.

Pediatric Blood Cancer 2004;43:606–609.

62. Chauvenet AR, Shashi V, Selsky C, et al. Vincristine-induced

neuropathy as the initial presentation of charcot-marie-tooth

disease in acute lymphoblastic leukemia: A Pediatric Oncology

Group study. Am J Pediatr Hematol Oncol 2003;25:316–320.

63. Kalfakis N, Panas M, Karadima G, et al. Hereditary neuropathy

with liability to pressure palsies emerging during vincristine

treatment. Neurology 2002;59:1470–1471.

64. Naumann R, Mohm J, Reuner U, et al. Early recognition of

hereditary motor and sensory neuropathy type 1 can avoid life-

threatening vincristine neurotoxicity. Br J Haematol 2001;115:

323–325.

65. Hildebrandt G, Holler E, Woenkhaus M, et al. Acute deterioration

of Charcot-Marie-Tooth disease IA (CMT IA) following 2 mg of

vincristine chemotherapy. Ann Oncol 2000;11:743–747.

66. Mercuri E, Poulton J, Buck J, et al. Vincristine treatment revealing

asymptomatic hereditary motor sensory neuropathy type 1A. Arch

Dis Child 1999;81:442–443.

67. Olek MJ, Bordeaux B, Leshner RT. Charcot-Marie-Tooth disease

type I diagnosed in a 5-year-old boy after vincristine neurotoxicity,

resulting in maternal diagnosis. J Am Osteopath Assoc 1999;99:

165–167.

68. Ajitsaria R, Reilly M, Anderson J. Uneventful administration of

vincristine in Charcot-Marie-Tooth disease type 1X. Pediatric

Blood Cancer 2008;50:874–876.

69. Porter CC, Carver AE, Albano EA. Vincristine induced peripheral

neuropathy potentiated by voriconazole in a patient with previously

undiagnosed CMT1X. Pediatric Blood Cancer 2009;52:298–300.

70. Kivisto KT, Kroemer HK, Eichelbaum M. The role of human

cytochrome P450 enzymes in the metabolism of anticancer agents:

Implications for drug interactions. Br J Clin Pharmacol 1995;40:

523–530.

71. Sathiapalan RK, El-Solh H. Enhanced vincristine neurotoxicity

from drug interactions: Case report and review of literature. Pediatr

Hematol Oncol 2001;18:543–546.

72. Sathiapalan RK, Al-Nasser A, El-Solh H, et al. Vincristine-

itraconazole interaction: Cause for increasing concern. Am J

Pediatr Hematol Oncol 2002;24:591.

73. Mantadakis E, Amoiridis G, Kondi A, et al. Possible increase of the

neurotoxicity of vincristine by the concurrent use of posaconazole

in a young adult with leukemia. Am J Pediatr Hematol Oncol

2007;29:130.

74. Takahashi N, Kameoka Y, Yamanaka Y, et al. Itraconazole oral

solution enhanced vincristine neurotoxicity in five patients with

malignant lymphoma. Intern Med 2008;47:651–653.

75. Bermudez M, Fuster JL, Llinares E, et al. Itraconazole-related

increased vincristine neurotoxicity: Case report and review of

literature. Am J Pediatr Hematol Oncol 2005;27:389–392.

76. Ariffin H, Omar KZ, Ang EL, et al. Severe vincristine neurotoxicity

with concomitant use of itraconazole. J Paediatr Child Health 2003;

39:638–639.

77. Jeng MR, Feusner J. Itraconazole-enhanced vincristine neuro-

toxicity in a child with acute lymphoblastic leukemia. Pediatr

Hematol Oncol 2001;18:137–142.

78. Kamaluddin M, McNally P, Breatnach F, et al. Potentiation of

vincristine toxicity by itraconazole in children with lymphoid

malignancies. Acta Paediatr 2001;90:1204–1207.

79. Worth LJ, Blyth CC, Booth DL, et al. Optimizing antifungal drug

dosing and monitoring to avoid toxicity and improve outcomes

in patients with haematological disorders. Intern Med J 2008;38:

521–537.

80. Antoniou T, Tseng AL. Interactions between antiretrovirals and

antineoplastic drug therapy. Clin Pharmacokinet 2005;44:111–145.

81. Worth LJ. RE: Azoles and Vincristine Dosing Recommendations.

Personal communication, 2009.

82. Merck & Co., Inc. Emend (aprepitant) prescribing information.

Whitehouse Station, NJ: Merck & Co., Inc.; 2008.

83. Villikka K, Kivisto KT, Maenpaa H, et al. Cytochrome P450-

inducing antiepileptics increase the clearance of vincristine in

patients with brain tumors. Clin Pharmacol Ther 1999;66:589–

593.

84. Balayssac D, Cayre A, Ling B, et al. Vincristine-induced neuro-

pathy in the rat is not modified by drug-drug interactions with the

P-glycoprotein inhibitor verapamil. Chemotherapy 2008;54:336–

342.

85. Muthuraman A, Jaggi AS, Singh N, et al. Ameliorative effects of

amiloride and pralidoxime in chronic constriction injury and

Pediatr Blood Cancer DOI 10.1002/pbc

1186 Moore and Pinkerton

Page 8: Vincristine: Can its therapeutic index be enhanced?

vincristine induced painful neuropathy in rats. Eur J Pharmacol

2008;587:104–111.

86. Bujalska M, Tatarkiewicz J, Gumulka SW. Effect of

bradykinin receptor antagonists on vincristine- and streptozoto-

cin-induced hyperalgesia in a rat model of chemotherapy-

induced and diabetic neuropathy. Pharmacology 2008;81:158–

163.

87. Kamei J, Nozaki C, Saitoh A. Effect of mexiletine on vincristine-

induced painful neuropathy in mice. Eur J Pharmacol 2006;536:

123–127.

88. Sweitzer SM, Pahl JL, DeLeo JA. Propentofylline attenuates

vincristine-induced peripheral neuropathy in the rat. Neurosci Lett

2006;400:258–261.

89. Wang M, Wu Y, Culver DG, et al. The gene for slow Wallerian

degeneration (Wld(s)) is also protective against vincristine neuro-

pathy. Neurobiol Dis 2001;8:155–161.

90. Watanabe M, Tsukiyama T, Hatakeyama S. Protection of

vincristine-induced neuropathy by WldS expression and the

independence of the activity of Nmnat1. Neurosci Lett 2007;411:

228–232.

91. Pan YA, Misgeld T, Lichtman JW, et al. Effects of neurotoxic and

neuroprotective agents on peripheral nerve regeneration assayed

by time-lapse imaging in vivo. J Neurosci 2003;23:11479–

11488.

92. Koeppen S, Verstappen CC, Korte R, et al. Lack of neuroprotection

by an ACTH (4–9) analogue. A randomized trial in patients treated

with vincristine for Hodgkin or non-Hodgkin lymphoma. J Cancer

Res Clin Oncol 2004;130:153–160.

93. Veerman AJ, Hahlen K, Kamps WA, et al. High cure rate with a

moderately intensive treatment regimen in non-high-risk childhood

acute lymphoblastic leukemia. Results of protocol ALLVI from the

Dutch Childhood Leukemia Study Group. J Clin Oncol 1996;14:

911–918.

94. Pinkel D, Hernandez K, Borella L, et al. Drug dosage and remission

duration in childhood lymphocytic leukemia. Cancer 1971;27:

247–256.

95. Davies SM. Pharmacogenetics, pharmacogenomics and personal-

ized medicine: Are we there yet? Hematology/the Education

Program of the American Society of Hematology American

Society of Hematology Education Program 2006; pp. 111–117.

Pediatr Blood Cancer DOI 10.1002/pbc

Vincristine: Can Its Therapeutic Index Be Enhanced? 1187