Sie sind auf Seite 1von 18

Review Article

Cisplatin: a review of toxicities and


therapeutic applications
K. Barabas, R. Milner, D. Lurie and C. Adin
Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida,
Gainesville, FL, USA

Abstract
Cisplatin is a platinum chemotherapeutic used in a variety of malignancies. The antineoplastic activ-
ity occurs from DNA cross-links and adducts, in addition to the generation of superoxide radicals.
Nephrotoxicity is the most well-known and potentially most clinically signicant toxicity. Unfortu-
nately, the mechanism for cisplatin nephrotoxicity has not been completely elucidated; however,
many theories have been developed. Other toxicities include gastrointestinal, myelosuppression,
ototoxicity and neurotoxicity. Saline diuresis is currently the most accepted way to prevent cisplatin
nephrotoxicity. Research has focused on pharmaceuticals and enzyme/molecular alterations as
alternatives to long-term diuresis. No agents have currently been identied that can protect from all
toxicities. Cisplatin has shown activity against osteosarcoma, transitional cell carcinoma, squamous
cell carcinoma (SCC), melanoma, mesothelioma, carcinomatosis and germinal cell tumours in the
Keywords dog. In the cat, cisplatin cannot be utilized because of fulminant pulmonary oedema that occurs at
chemotherapy, cisplatin,
nephrotoxicity,
standard doses. Intralesional cisplatin has been utilized in horses for the treatment of SCC and
osteosarcoma sarcoids.

Introduction
(OSA), transitional cell carcinoma (TCC), intrale-
Cisplatin is one of the most potent chemotherapy sional therapy and radiation sensitization.
agents used in human and veterinary medicine. Its Since cisplatins development, research has centred
use in veterinary medicine began more than two on mitigating nephrotoxicity to allow cisplatin to be
decades ago and was prompted by the success of delivered at therapeutic doses without adversely af-
cisplatin in treating human malignancies. Unfortu- fecting the kidneys. Recent studies have discovered
nately, cisplatin administration was associated with new protocols, compounds, enzymes and molecular
numerous side-effects including nephrotoxicity, alterations that reduce the nephrotoxicity of cisplatin.
severe nausea and vomiting, myelosuppression, This review will focus on cisplatins chemistry,
ototoxicity and neurotoxicity. Of these, the most pharmacokinetics, pharmacodynamics, mecha-
clinically signicant and common toxicity is neph- nisms of resistance, toxicity, prevention of nephro-
Correspondence address: rotoxicity. Despite the nephrotoxicity, many veter- toxicity and use in animals. Also included is a
Rowan Milner inary oncologists are of the opinion that cisplatin is section on the other platinum drugs that have been
Department of Small Animal
more potent than its other platinum counterparts used in veterinary medicine to date.
Clinical Sciences
Veterinary Medical Center with regard to its antineoplastic activity; however,
University of Florida there is no current scientic basis for this opinion
PO Box 100126, Gainesville Chemistry
in veterinary oncology. This has resulted in its con-
FL 32610-0126, USA
e-mail: milnerr@mail. tinued use throughout veterinary hospitals around Cisplatin was inadvertently discovered while study-
vetmed.u.edu the world for malignancies such as osteosarcoma ing the growth characteristics of Escherichia coli.1

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd 1


2 K. Barabas et al.

Initially, researchers observed that platinum com- levels corresponded with nephrotoxicity.8 Plasma
pounds exhibited antibacterial properties, and platinum levels may serve as a marker for risk of
subsequently, it was discovered that they also po- nephrotoxicity in certain patients.
ssessed antineoplastic properties.1,2 The molecular Urinary levels of platinum in the dog increase
structure of cisplatin comprises a central platinum rapidly after administration, with 60% of the dose
atom surrounded by two chlorine atoms and two recovered in the urine within the rst 4 h and 76%
ammonia groups in a cis conguration.3 Other of the administered dose recovered by 48 h after
platinum compounds have the same core platinum treatment.7,9 Only small amounts of platinum were
compound, and cis conguration; however, their detected in bile, suggesting minimal faecal excre-
leaving groups are different (Fig. 1).4 The bond tion.7 Free platinum clearance has been shown to
angles for the platinum core are xed, resulting in exceed the creatinine clearance by 156%, suggest-
DNA bending to accommodate the structure of the ing that in addition to excretion by ltration, cispl-
drug.4 atin or a metabolite is secreted by the kidney.5 It is
thought that secretion involves active accumula-
tion of secreted substances in the renal cells and
Pharmacokinetics
passive transport into the tubule of the lumen.10
Plasma platinum has been shown to be highly pro- The pars recta of the proximal tubule is the most
tein bound.5 Most cisplatin present in the cell is active site of secretion and also the most damaged
found in the cytosol and is not protein bound.6 site of the kidney during cisplatin nephrotoxicity.5
Cisplatins clearance in the dog is biphasic in na- Renal accumulation of platinum is dependent on
ture, with a rapid phase half-life of 22 min and a the presence of normal oxygen utilization and the
slow phase half-life of 5 days.7 Signicant amounts organic base transport system.11 Thus, concurrent
of platinum are still detectable in plasma 12 days administration of drugs known to be transported
after intravenous injection.7 A study performed in by this system can signicantly reduce cisplatin up-
humans demonstrated that peak plasma platinum take in the kidney.11

Figure 1. Two-dimensional structures for cisplatin, carboplatin, oxaliplatin and lobaplatin. While the core structure (Pt) is
the same for each drug, the leaving groups are different for each compound.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 3

Cisplatin is initially distributed to all tissues; action does not occur and the drug remains
however, in the rst hour, it tends to accumulate in neutral.13 The neutral form is believed to be bio-
the kidney, liver, muscle and skin.7 The localization logically inactive.14 Intracellular uid has approxi-
in the kidney and liver is protracted, with high re- mately one-thirteenth the chloride concentration
nal tissue concentrations present as long as 12 days of extracellular uid, and it is under these condi-
after treatment in the dog.7 The highest tissue tions that the aquation reaction proceeds, leading
platinum concentrations occur in those tissues to eventual DNA damage.13 The primary effect
where the drug exerts its most potent antineoplas- produced by cisplatin in cancerous cells is inhibi-
tic activity, such as the ovary and uterus.7 It is tion of DNA synthesis.15,16 The ability to inhibit
thought that the presence of tumour may alter the DNA synthesis occurs at much lower doses than
toxicity and pharmacokinetics of drugs.12 One that necessary to inhibit RNA and protein synthe-
study in tumour-bearing rats showed that the dis- sis.15 DNA damage induced by cisplatin is similar
tribution half-time was longer for the tumour- to that caused by alkylating agents.17 With aqua-
bearing rats than for their controls, while the tion of the platinum compound, the two chloride
terminal elimination half-time was the same for groups are replaced with water and will bind to two
both groups.12 Based on this study, it is unlikely sites in DNA.4 Generally, if the two sites are on the
that tumour presence would alter toxicity or other same DNA strand, the lesion is referred to as a
distribution-dependent drug parameters.12 DNA adduct and if the sites are on different strands,
the lesion is referred to as a DNA cross-link.4 Cis-
platin has been noted to bind to all DNA bases but
Pharmacodynamics
has a preference for the N-7 positions of adenine
Cisplatin is activated by an aquation reaction in- and guanine because of the high nucleophilicity of
volving the exchange of the two chloride leaving the N-7 sites of these purine bases (Fig. 2).4,13,18
groups with water or hydroxyl ligands.13 When a Cisplatin forms bifunctional adducts >90% of the
high concentration of chloride is present, as in iso- time with cross-links being <2% of the lesions
tonic saline or extracellular uid, the aquation re- formed.4 These adducts and cross-links inhibit

Figure 2. Aquation reaction and adduct formation at the N-7 position of guanine on two sites of DNA. These adducts
result in DNA damage, resulting in cell kill.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
4 K. Barabas et al.

DNA template replication in mammalian cells.19 (MMR) mediates apoptosis in response to cispla-
DNA cross-links and adducts increase with time tin.3032 Defects in MMR result in altered cell sensi-
after the drug is removed and are repaired slowly.20 tivity to cisplatin, most likely resulting in greater
In vitro studies have also indicated that interaction resistance.4 Reports indicate that where alterations
between the cisplatin molecule and the DNA may in MMR exist, concurrent enhancement of the
contribute to the generation of superoxide radicals, activity of NER exists.4
causing further toxicity to cancer cells.21,22

Toxicities
Mechanisms of resistance
Nephrotoxicity
Four generic pathways for cisplatin resistance have
been uncovered. They include altered cellular ac- Cisplatin is associated with several systemic toxici-
cumulation, cytosolic inactivation of cisplatin, ties but is most frequently associated with nephro-
DNA repair and altered apoptosis (Fig. 3).4 In vitro toxicity. Cisplatin-induced nephrotoxicity occurs
studies have described active efflux particularly in a number of species including mice, rats, dogs
as mediated by CuH transporters, ATP7A and and humans. It is estimated that 2836% of human
ATP7B, and other less well-dened systems.4 Co- patients receiving an initial dose of 50100 mg m2
valent binding of proteins or peptides with in- of cisplatin develop acute renal failure.33,34 Cispla-
creased levels of sulfhydryl groups to cisplatin may tin nephrotoxicity is dose and duration of treat-
confer cellular resistance.4 These compounds in- ment dependent and is enhanced by the use of
clude glutathione (GSH) and metallothionein.2326 other nephrotoxins such as aminoglycosides.35,36
MRP2 (multidrug resistance-associated protein 2) Most patients who develop some degree of renal
may also play a role in cisplatin resistance by re- dysfunction never fully recover.37 However, one
moving the cisplatinGSH complex from the study evaluating the long-term renal effects of cis-
cells.27 PlatinumDNA repair occurs by the nucle- platin in human patients showed that renal dys-
otide excision repair (NER) and NER is increased function may not be progressive provided further
in cisplatin-resistant cells.4,28,29 Mismatch repair insult is avoided. In this study, an initial increase in

Figure 3. Four pathways exist for cisplatin resistance. They include: (1) decreased cellular accumulation, (2) inactivation of
the drug, (3) DNA repair and (4) prevention of apoptosis.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 5

creatinine and a decrease in glomerular ltration where can or the levels of DNA adducts in the pars
rate (GFR) and renal plasma ow were noted di- recta are lethal, whereas those produced in other
rectly after treatment, but these levels remained segments are not.42,43 Interestingly, recent studies
stable for up to 1224 months after discontinuing have related cisplatin administration to alterations
cisplatin treatment.38 in the cell cycle. Cells in the kidney enter the cell
The morphologic alterations in the kidney at- cycle after cisplatin administration and genes for
tributed to cisplatin administration occur in the the p21 and 14-3-3 cell cycle inhibitors are simul-
pars recta of the proximal tubule situated in the taneously upregulated.4446 Mice with a deleted p21
outer stripe of the medulla.39 Histological changes gene were more sensitive to cisplatin injury.46 Price
are consistent with both apoptosis and necrosis.40 et al. showed that the addition of p21 by adenoviral
One of the earliest histopathological changes noted vector and the pharmacological inhibition of cyclin-
is the swelling of mitochondria.14 Most of the path- dependent kinases by roscovitine, both protected
ological changes start 3 days after cisplatin admin- kidney cells from cisplatin-induced nephrotoxicity
istration, including clumped nuclear chromatin, in vitro.47 p21 inhibits caspase activation, which is
increased number of cytoplasmic vesicles, focal discussed later as a mechanism of apoptosis in the
loss of microvillus brush border and completely renal tubule.47
necrotic cells sloughed in the tubular lumen.39 The As platinum drugs are similar to other heavy met-
most severe damage is seen 5 days after cisplatin als such as mercury, another possible mechanism for
administration and consists of widespread tubular nephrotoxicity incorporates the known mechanism
necrosis in the pars recta, desquamation of necrotic of nephrotoxicity by other heavy metals.48 This toxic-
epithelia cells resulting in a denuded basement ity occurs as a result of the binding of cisplatin to sulf-
membrane, necrotic cells and debris in the tubular hydryl (SH) groups in the kidney that are necessary
lumen and the changes seen in 3 days after cisplatin for enzyme function and depletion of intracellular
administration for non-necrotic cells.39 By 7 days glutathione.48,49 The decrease in SH groups occurred
after cisplatin administration, extensive regenera- before the rise in blood urea nitrogen (BUN) and cre-
tion in the pars recta is noted with necrotic cells atinine and was not seen with acute renal failure
still present.39 caused by glycerol, another potent nephrotoxin.49
Research into the mechanism of cisplatin neph- Proximal tubular cell death was initially believed
rotoxicity is an important step in developing meth- to occur mostly by necrosis. However, another
ods for renal protection. One theory involves DNA mechanism of proximal tubule cell death is apop-
cross-links and the position in the cell cycle. It was tosis. A previous study showed that the type of cell
thought that cisplatinDNA cross-links could be death was concentration dependent, with high
the cause of cytotoxicity in the renal cell, but the concentrations of cisplatin leading to necrosis and
proximal tubule cells selectively killed by cisplatin low concentrations causing apoptosis.50 Reactive
are relatively quiescent and therefore should not be oxygen species (ROS) and mitochondria are
as sensitive to the toxicity of DNA damaging thought to play a role in the apoptotic cascade.51
agents.41 However, there is a fall in DNA turnover Mitochondrial dysfunction occurs early in cisplatin-
that precedes necrosis in the proximal tubule and induced renal tubular toxicity and is potentially
the later increase in DNA turnover in those cells mediated by ROS.52,53 It was shown in a previous
coincides with the timeline of regeneration.42 Both study that overexpression of manganese superox-
the outer cortex and the outer stripe of the outer ide dismutase, an anti-oxidant enzyme found in
medulla (pars recta) have decreased DNA synthesis mitochondria, protected renal epithelial cells in vitro
1 day after cisplatin administration; yet, only the from cisplatin toxicity.54 In vitro studies per-
cells in the pars recta undergo necrosis.42 Three formed on renal proximal tubule cells examined
possibilities have been considered regarding this the role of caspases and p53 in apoptosis related
theory including inhibition of DNA synthesis is to cisplatin. Lau et al. showed that caspase 3 was
irrelevant to cytotoxicity in the kidney, cells in the activated in vitro in response to cisplatin but the
pars recta cannot repair the damage as cells else- initiators of the activation were not found.55 The

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
6 K. Barabas et al.

tumour suppressor gene p53 is activated in re- decreased after cisplatin administration.38,42,68 Ini-
sponse to DNA damage, alterations in the cell cycle tially, it was thought that the reninangiotensin
and hypoxia.56 Another study found that 50% of system may play a role in cisplatin-induced acute
cisplatin-induced renal proximal tubular cell apop- renal failure, although experimental studies failed
tosis was mediated by p53 and that p53 activates to conrm this hypothesis.42,69 In rats, decreases in
caspase 3 independent of other caspases or mito- GFR are related to afferent vasoconstriction and
chondrial dysfunction.57 The other 50% is medi- possibly an altered ultraltration coefcient, both
ated by additional mechanisms independent of p53 of which occur before evidence of tubular obstruc-
and caspases 3, 8 and 9.57 Interestingly, a different tion.70 It should be restated that histopathologi-
in vitro study performed by Xiao et al. showed that cally, the glomerulus is minimally affected by
when a caspase inhibitor that has no effect on p53 cisplatin.
was applied to renal tubular cells, cisplatin-induced
apoptosis did not occur.58
Protective measures for nephrotoxicity
Recent studies in rats and mice have shown that
the nephrotoxicity of cisplatin can be blocked by
Saline diuresis
inhibiting either of two enzymes expressed in
proximal tubules, gamma-glutamyl transpeptidase The most common protocol for administering cis-
(GGT) or cysteine-S-conjugate beta-lyase.5961 This platin consists of pre- and posthydration with con-
suggested that metabolic activation of cisplatin to a current saline diuresis (Table 1). There are two
nephrotoxin occurred in the kidney.41 For this re- common short-term diuresis protocols utilized in
action to occur, cisplatin must form a conjugate the dog. The incidence of nephrotoxicity was simi-
with glutathione, which has been shown to occur lar between the studies, and survival times for the
spontaneously in solution.62,63 Selective inhibition dogs developing nephrotoxicosis were similar to
of each enzyme resulted in a decrease in toxicity, those that did not develop nephrotoxicosis in both
in vitro.41 Interestingly, conjugation of cisplatin with studies.71,72 The maintenance of adequate hydra-
glutathione reduces cisplatin cross-links with DNA, tion is important for decreasing nephrotoxicity,
resulting in decreased toxicity to dividing cells, but the mechanism of protection is unknown.
suggesting decreased antitumour activity.64 Also,
GGT expression in tumours has been shown to de-
Diuretics
crease the antitumour activity of cisplatin.65 These
conicting reports show that there are many areas Other methods for decreasing the nephrotoxicity
regarding mechanisms of cisplatin-induced apop- of cisplatin include mannitol or furosemide ad-
tosis and necrosis that still need to be investigated. ministration, although the latter is not currently
The physiologic alterations seen with cisplatin recommended in the dog. The exact mechanism
are relatively consistent. Renal failure is gradual behind diuretics ameliorating cisplatin toxicity is
and usually occurs 35 days after administration.66 unknown, but postulated mechanisms include ac-
Polyuria may occur because of a reduction in normal celerating the passage of cisplatin through the renal
corticalpapillary solute gradient in association tubules by increased urinary excretion, reversing
with a failure to recycle urea.67 Whole kidney GFR, the osmotic gradient in tubules by mannitol and
single nephron GFR and renal plasma ow are all blocking of sodium and water reabsorption by

Table 1. Canine diuresis protocols when administering cisplatin. These protocols utilized 70 mg m2 given with 0.9% NaCl
i.v. over 20 minutes

Duration of Fluid rate Length of diuresis Length of diuresis Incidence of


diuresis (h) (mL kg1 h1) before cisplatin (h) after cisplatin (h) nephrotoxicity (%) Reference

4 25 3 1 7.8 72
6 18.3 4 2 6.6 71

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 7

furosemide.73 It has also been suggested that these side-effects and ototoxicity have limited the clini-
diuretics may also attenuate cisplatin nephrotoxic- cal application of this drug.82 Probenecid is thought
ity, reducing the concentration of platinum in the to partially inhibit platinum renal secretion and
urine.9,74 However, in the study by Pera et al., it was subsequently decreases the platinum concentra-
noted that while diuretic administration signicantly tion in the renal tubules, decreasing nephrotoxicity
improved renal function, some degree of tubular without affecting cisplatins antitumour activity.83
necrosis was still present.74 Another study showed This drug is non-toxic, inexpensive and readily
that neither mannitol nor furosemide was superior available but failed to protect against the other
to the other in reducing nephrotoxicity in the human side-effects of cisplatin such as myelosuppression,
patients involved.73 However, there have been con- gastrointestinal toxicity and ototoxicity.83 Sodium
icting studies comparing hydration with or with- thiosulphate is an anti-oxidant in the thiol fam-
out mannitol. In one study, mannitol ameliorated ily.84 It is used most commonly in conjunction with
nephrotoxicity better than hydration alone.75 The intracavitary cisplatin to reduce toxicity and allows
conicting study used the same dose of cisplatin the dose of cisplatin to be delivered to be as high as
but stated that there was no difference between 270 mg m2.85 In a recent study performed on a rat
groups receiving mannitol and hydration or hydra- model, sodium thiosulphate was found to provide
tion alone.76 In spite of these conicting reports, protection from cisplatin ototoxicity when deliv-
administration of mannitol or furosemide along ered at 48 h after cisplatin but was not consistently
with continuous saline diuresis has become stan- protective against nephrotoxicity.86 Procainamide,
dard practice when using cisplatin chemotherapy an anti-arrhythmic agent, has also been shown to
in human cancer patients. protect against cisplatin nephrotoxicity without al-
tering its antitumour effects.87 Procainamide and
cisplatin form a complex that increases the amount
Hypertonic saline
of platinum bound to DNA and may prevent me-
Protection from nephrotoxicity was also seen when tabolism of cisplatin to a nephrotoxin by GGT
cisplatin was dissolved in a hypertonic NaCl solu- through the formation of a cisplatinglutathione
tion (4.5%) relative to distilled water with no effect complex.41,88 Methimazole, an antithyroid drug,
on the antitumour action of cisplatin.77 It is postu- was given intraperitoneally 30 min prior and 4 h af-
lated that the presence of the high concentration of ter cisplatin infusion without saline prehydration
NaCl in the vehicle was great enough to force the to normal dogs and was found to signicantly de-
aquation reaction far to the left, thus favouring the crease nephrotoxicity.89 Methimazole is thought to
presence of the parent cis molecule, decreasing bind- exert an anti-oxidative effect to protect the kidney,
ing to plasma proteins and tissue-binding sites.77 but it is unknown whether this compound affects
cisplatin tumouricidal activity.89 Liposome encap-
sulation of cisplatin has also been proven to allow
Pharmaceuticals
an increase in dose of cisplatin that can be safely
Additional drugs have been administered in con- administered without increasing the nephrotoxicity
junction with cisplatin to reduce nephrotoxicity. in dogs and cats without concurrent diuresis.9092
Amifostine (WR-2721) is an SH-containing com-
pound that when injected before cisplatin in rats
Enzymatic and molecular alterations
decreased nephrotoxicity by a factor of 1.7 without
inhibiting its antitumour effect.7880 Diethyldithio- Reduction of nephrotoxicity has also been associ-
carbamate is a chelating agent that potentially re- ated with some enzymes or agents that control or
moves platinum bound to renal tubules.81 However, prevent the formation of free radicals. One study
several side-effects (hypertension, agitation, ush- demonstrated a decrease in cisplatin nephrotoxic-
ing and diaphoresis) that required patients to re- ity in rats treated with a superoxide dismutase
ceive sedation during administration and the mimetic, orgotein.93 Another rat model study used
failure of the drug to ameliorate gastrointestinal N-acetylcysteine, an anti-oxidant, delivered at

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
8 K. Barabas et al.

400 mg kg1 i.v. 15 min prior to cisplatin injection for this is still unclear, but studies in rats suggest
and found that treated rats had normal BUN and that abnormal magnesium excretion may be be-
creatinine and histologically normal kidneys 3 days cause of a defect in magnesium transport in juxta-
after injection.86 Many studies investigating the medullary nephrons or collecting ducts.104 When
mechanism of cisplatin nephrotoxicity have used clinical manifestations of hypomagnesaemia such as
agents such as caspase inhibitors and anti-oxidants neuromuscular, CNS and cardiac function abnor-
to view their role in apoptosis of renal tubular malities occur, they are usually seen with serum lev-
cells.47,54,58 Also, upregulation of p21 and manganese els less than 1 mEq L1, and parenteral replacement
superoxide dismutase is considered as a potential of magnesium sulphate should be administered.34
future therapy, as studies carried out in vitro have Hypomagnesaemia is usually complicated by
demonstrated that upregulation of these two genes hypocalcaemia that is probably secondary to di-
resulted in protection against cisplatin nephrotoxic- minished parathyroid hormone release and/or
ity.47,54 Unfortunately, many of these are in vitro end-organ resistance to parathyroid hormone in-
studies on kidney tubule cells and effects in higher duced by hypomagnesaemia.105,106 Hypocalcaemia
mammals, and antitumour effect is still unknown. resolves when magnesium is replaced and is unre-
Another recent development has been with the sponsive to calcium replacement alone.34
endogenous enzyme heme oxygenase-1 (HO-1).
HO-1 is an inducible enzyme that degrades heme
Gastrointestinal toxicity and
and produces carbon monoxide (CO), iron and
myelosuppression
biliverdin. Biliverdin is reduced to bilirubin in vivo
and is a powerful anti-oxidant.94,95 Carbon mon- Gastrointestinal toxicity and myelosuppression ap-
oxide possesses vasodilatory, anti-inammatory pear to be associated with the death of the rapidly
and anti-apoptotic properties.9699 HO-1 upregula- dividing cells in the lining of the gastrointestinal
tion occurs after cisplatin administration and upreg- tract and in the bone marrow. Cisplatin also acti-
ulation protects against cisplatin nephrotoxicity.100 vates the chemoreceptor trigger zone to induce
Tayem et al. recently showed that a water-soluble vomiting.3 While several anti-emetics, including
carbon monoxide-releasing molecule protected renal butorphanol, metoclopramide, dolasetron, ondan-
tubular cells from cisplatin injury in vitro and setron, and chlorpromazine, have been recom-
in vivo in rats.101 Again, as with other recent studies mended prior to, during and after cisplatin infusion
mentioned previously, it is unknown whether to decrease nausea and vomiting, clinical efcacy
HO-1 interferes with cisplatins antitumour effect. data exist only for the use of butorphanol for this
Recently, in our laboratory, we have reported that indication in the dog.107,108 Recently, the neuroki-
hyperbilirubinaemia ameliorates nephrotoxicity in nin 1 receptor antagonist, maropitant (Cerenia)
rats receiving cisplatin.102 has been shown in randomized clinical trials to be
highly effective for preventing and treating cisplatin-
induced emesis and is the only anti-emetic ap-
Hypomagnesaemia and hypocalcaemia
proved for use in dogs for this use in both Europe
Other physiologic changes related to cisplatin ad- and the USA.109
ministration include hypomagnesaemia and hypo-
calcaemia. Hypomagnesaemia has been reported to
Ototoxicity
occur in more than half of human patients receiving
cisplatin chemotherapy.103 The persistent excretion Ototoxicity has been reported more commonly
of magnesium in the presence of declining magne- in human patients than in the dog, likely because
sium levels suggests that the hypomagnesaemia is of our inability to determine this clinical change
because of a renal defect in magnesium reabsorp- consistently in the latter species. Ototoxicity has
tion.103 This is not necessarily associated with overt been observed in 790% of human patients receiv-
renal insufciency and may be a more common ing doses of up to 120 mg m2 per course.110,111
manifestation than renal failure.103 The mechanism Hearing loss is in the high-frequency range and is

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 9

dose related, cumulative and frequently irrevers- intra-arterial, intramedullary, intralesional and in-
ible.112,113 Concurrent cranial irradiation enhances tracavitary routes. Two short-term diuresis proto-
the ototoxicity.82 In one study with high-dose cis- cols have been utilized in the dog for intravenous
platin, in spite of signicantly lower hearing levels, administration and have been discussed previously
no human patient suffered a disabling hearing loss, (Table 1).
requiring a hearing aid, and the use of hypertonic Cisplatin is most commonly used as a single agent;
saline and vigorous hydration was not found to however, it is occasionally used as part of combina-
minimize ototoxiciy.114 While humans manifest tion chemotherapy with doxorubicin to treat OSA.
this toxicity in hearing a high-pitched tinnitus Median survival times in dogs treated with cisplatin
(ringing in ears) and hearing sounds differently, as the sole chemotherapeutic agent range from 262
small animals may express this as an inappropriate to 413 days.118122 Various combination protocols
response or an unusually strong response to an au- have been investigated. Doxorubicin (30 mg m2 i.v.
ditory stimulus, such as hyperactivity or excessive on day 1) and cisplatin (60 mg m2 i.v. on day 21) re-
barking.115 The mechanism for this toxicity is un- peated for two treatment cycles resulted in median
clear but may involve spontaneously recruiting ad- survival times of 300 days.123 A more recent study
jacent neurons, aberrant cochlear uid currents or investigated the use of doxorubicin and cisplatin ad-
undermodulation of membrane movement.115 ministered within 24 h of each other.124 Cisplatin
(50 mg m2) was administered i.v. followed by doxo-
rubicin (15 mg m2) 24 h later with the intent of
Neurotoxicity
completing four treatment cycles.124 Median sur-
Neurotoxicity is described as a peripheral neuropa- vival times were equivalent to those in studies where
thy and usually develops in human patients that cisplatin and doxorubicin were administered 3 weeks
receive a cumulative dose of 400 mg m2 or apart at 300 days, but signicant toxicity was en-
higher.116 In studies using high-dose cisplatin with countered with this protocol.124 Renal toxicity was
protective measures, mild to moderate paresthesias present in 11% of patients, which is higher than the
have occurred in some human patients.82,83 reported incidence in the diuresis studies utilizing
cisplatin as a sole treatment agent.124 Gastrointesti-
nal toxicity and myelosuppression were comparable
Syndrome of inappropriate secretion of
to those in larger studies utilizing only cisplatin.124
antidiuretic hormone 126
The use of STEALTH liposome-encapsulated
Syndrome of inappropriate secretion of antidi- cisplatin versus carboplatin has found that while the
uretic hormone (SIADH) has been reported in use of the STEALTH cisplatin allowed safe adminis-
the human literature with the administration of tration of ve times the maximally tolerated dose of
cisplatin and other cytotoxic drugs.117 SIADH is free cisplatin, this did not translate into prolonged
characterized by hyponatraemia with concurrent disease-free or overall survival. STEALTH liposomes
hypo-osmolality of the serum; continued renal received their name by avoiding uptake by the
excretion of sodium; no clinical evidence of volume mononuclear phagocytic system.127
depletion; urine osmolality greater than that ap- Other methods of administration of cisplatin for
propriate for concurrent osmolality of serum and the treatment of OSA that have been investigated
normal function of the kidneys, suprarenal glands include intralesional (with implants or injection),
and thyroid glands.117 intra-arterial and intramedullary administration.
Cisplatin-containing implants have been utilized
in limb-spare procedures for additional local con-
Cisplatin use in animals
trol.128 Dogs receiving cisplatin implants were
53.5% less likely to develop local recurrence than
Dogs
the control groups, although this effect did not
Cisplatin has been used as a systemic or local reach statistical signicance.128 Intramedullary cis-
chemotherapy agent in dogs via intravenous, platin (60 mg m2 over 20 min) administered with

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
10 K. Barabas et al.

a Jamshidi biopsy needle inserted into the tumour of 363 days, which was longer than the previously
provided effective local control in two of four dogs reported survival times for surgery or radiation
unable to undergo amputation or a limb-spare therapy alone.138 It should be noted that the dogs in
procedure.129 this study had small initial tumour size and no
Cisplatin has also been utilized for TCC and lymph node or lung metastasis at the start, possibly
squamous cell carcinomas (SCC). At a dose of positively affecting survival times.138
50 mg m2 given i.v. every 28 days, cisplatin was Intralesional cisplatin has also become a possi-
found to have a palliative effect for dogs with SCC bility for local cisplatin administration. Implants
and TCC.130 No complete responses were observed containing cisplatin, viscous gel and a vasoactive
in this study, but partial remission and stable dis- modier have been used as primary treatment for
ease were observed in the majority of dogs without melanomas.139 Implants were injected until tu-
signicant toxicity noted.130 In another study in mour saturation was visualized, and these treat-
which cisplatin was given at 60 mg m2 i.v. every ments occurred weekly until complete tumour
3 weeks to dogs with a variety of malignant tu- resolution was observed.139 Seventy per cent of
mours that included TCC and SCC, an overall re- dogs had a >50% decrease in volume, and 55% of
sponse rate (complete or partial remission) of 19% these dogs had a complete response.139 Tumours
was observed.126 Those dogs demonstrating pro- that responded received a mean of 2.6 treat-
gressive disease did so by 42 days after the start of ments.139 The most common side-effect was local
treatment, suggesting that dogs should be evaluated necrosis seen in 85% of patients and was associated
at this time to determine response to treatment.126 with tumour response.139 Systemic toxicosis was
It is important to note that the combination of minimal with no dog exhibiting renal toxicosis.139
cisplatin and piroxicam (a non-steroidal anti- Patient survival was comparable to other forms of
inammatory agent also commonly used to treat local treatment such as radiation and surgery.139
TCC) cannot be recommended as the likelihood of Although there are no current published reports in
nephrotoxicity is greatly increased. dogs, intralesional cisplatin is also being utilized
The use of cisplatin concurrently with radiation for other tumours using sesame oil instead of the
therapy is a source of interest as cisplatin report- viscous gel described above.
edly enhances radiation-induced cell kill.131135 A Intracavitary cisplatin has also been evaluated
study performed on dogs with nasosinus carcino- in dogs. In one study, 50 mg m2 of cisplatin was
mas compared cobalt radiation alone with radia- administered every 28 days for a median of 2.5
tion in addition to cisplatin at 7.5 mg m2 i.v. bolus treatments to dogs with mesothelioma and carci-
before every other radiation treatment.136 The nomatosis.140 When using intracavitary chemo-
mean and median tumour control and survival therapy, the tumour is exposed by the capillary
times were not signicantly different between blood supply to a concentration equivalent to that
treatment groups; however, there was a trend to- achieved by i.v. administration, and the surface cell
ward longer tumour control and survival times in layers are exposed to a concentration that is 13
the cisplatin treatment group.136 Another study re- logs higher.141,142 Although intracavitary cisplatin
garding nasal tumours utilized an OPLA-Pt im- administration is a local method of chemotherapy
plant concurrently with radiation and found that delivery, signicant systemic absorption does oc-
the implant was clinically tolerable and yielded cur and the dose-limiting toxicity for intracavitary
comparable or perhaps improved survival times cisplatin is renal toxicity.140 Animals in this study
when compared with other published protocols.137 underwent diuresis, and 66.7% of dogs received
The use of radiation with cisplatin has also been additional treatment with sodium thiosulphate,
researched with regard to oral melanomas. In one preventing toxicity during the study.140 This
study, cisplatin 1030 mg m2 i.v. with diuresis or method of cisplatin delivery was associated with
carboplatin 90 mg m2 i.v. prior to 6 weekly 6-Gy palliation and control of malignant pleural and/or
radiation fractions was given.138 The use of low- abdominal effusion in ve of six dogs and pallia-
dose chemotherapy resulted in a median survival tion lasted 129 to greater than 807 days.140 Although

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 11

results for the previous study were promising, a cisplatin should be administered peri-operatively
more recent study found that the use of intracavi- or early in the postoperative period.150
tary carboplatin or mitoxantrone was just as effec-
tive for dogs with similar diseases.143 The ease of
Other platinum drugs
administration for carboplatin and mitoxantrone
is superior to cisplatin because diuresis is not re- The most commonly used alternative platinum
quired and minimal side-effects were noted, drug to cisplatin is carboplatin. Carboplatin has
suggesting that the use of cisplatin in intracavitary been shown to be less nephrotoxic than cisplatin
infusions will become obsolete in veterinary and can be administered without saline diuresis.
medicine.143 Carboplatin alone or in combination with doxoru-
bicin has been utilized to treat dogs with OSA with
survival times at 321 days and 320 days, respectively,
Cats which are similar to survival times achieved with
cisplatin alone or in combination with doxorubi-
Cisplatin is unable to be administered to cats be-
cin.151,152 However, in regard to treating TCC, car-
cause of its acute drug toxicity in this species. Cats
boplatin was not as effective as cisplatin in producing
receiving 60 mg m2 of cisplatin became dyspnoeic
a clinical response, although there was no direct
and died 4896 h after administration.144 Postmor-
comparison made.153 Other tumours in which car-
tem ndings included severe hydrothorax, pulmo-
boplatins use has been researched include malig-
nary oedema and mediastinal oedema.144 Lowering
nant melanoma, nasal tumours and anal sac
the dose of cisplatin to 40 mg m2 resulted in simi-
adenocarcinoma. As mentioned previously, carbo-
lar but less severe pulmonary changes, while de-
platin has additional uses as an intracavitary che-
creasing the dose to 20 mg m2 resulted in no
motherapeutic agent and as a radiosensitizer.138,143
pulmonary changes.144 The tumouricidal activity
Lobaplatin, another platinum analogue, was
of cisplatin alone at such doses is not known. Using
found to result in an 1-year survival fraction of
repetitive low dosing (10 mg m2, three times a
31.8% when administered every 3 weeks to dogs
week for 10 treatments), cisplatin use resulted in
with appendicular OSA.154 Clinical signs related to
reversible pulmonary oedema and renal insuf-
toxicosis were uncommon and usually were vomit-
ciency.145 The use of liposome-encapsulated cispla-
ing and depression.154 Unlike cisplatin, lobaplatin
tin has been investigated in cats. Studies with this
did not require pretreatment infusions.
form of cisplatin showed no renal or pulmonary
toxicity, but all cats had transient pyrexia and/or
lethargy, vomiting, inappetence and an acute infu- Summary
sion reaction prevented by administering atropine
Cisplatin has long been utilized in both human and
diphenhydramine.90,91 A different liposomal platinum
veterinary medicine. Cisplatin is still used widely in
analogue was evaluated in cats with SCC and found
many protocols in human patients affected with
to be an ineffective treatment.146
head and neck, lung and germ cell tumours, as well
as with OSA. The use of cisplatin in veterinary med-
icine is not as widespread, likely because of the re-
Horses
quirement of concurrent diuresis and the greater
Intratumoural cisplatin in oily emulsion has proven ease of using other platinum analogues that do not
efcacious in treatment of cutaneous tumours in require this step. Carboplatin, the most commonly
horses such as SCC and sarcoids.147,148 Intralesional used platinum analogue, is less nephrotoxic and has
cisplatin can be used alone for treatment of small recently become a more cost-effective alternative to
tumours or in combination with surgery for larger cisplatin. It is still likely that cisplatin will continue
tumours.149 Treatments are given at 2-week inter- to be utilized in veterinary medicine for treatment of
vals at a dose of 1 mg cisplatin for each cm3 of tissue OSA, intralesional chemotherapy for a variety of tu-
in the target eld.150 When surgery is performed, mours and as a sensitizer prior to radiation therapy.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
12 K. Barabas et al.

References 14. Litterst CL. Cisplatinum: a review, with special


reference to cellular and molecular interactions.
1. Rosenberg B, VanCamp L and Krigas T. Inhibition Agents and Actions 1984; 15: 520524.
of cell division in Escherichia coli by electrolysis 15. Harder HC and Rosenberg B. Inhibitory effects
products from a platinum electrode. Nature 1965; of anti-tumor platinum compounds on DNA,
205: 698699. RNA and protein syntheses in mammalian cells
2. Rosenberg B, VanCamp L, Trosko JE and Mansour in vitro. International Journal of Cancer 1970; 6:
VH. Platinum compounds: a new class of potent 207216.
antitumour agents. Nature 1969; 222: 385386. 16. Howle JA and Gale GR. Cis-
3. Page R, Matus RE, Leifer CE and Loar A. dichlorodiammineplatinum (II). Persistent and
Cisplatin, a new antineoplastic drug in veterinary selective inhibition of deoxyribonucleic acid
medicine. Journal of the American Veterinary synthesis in vivo. Biochemistry and Pharmacology
Medical Association 1985; 186: 288290. 1970; 19: 27572762.
4. Reed E. Cisplatin, carboplatin, and oxaliplatin. In: 17. Weiss RB and Poster DS. The renal toxicity of
Cancer Chemotherapy and Biotherapy Principles cancer chemotherapeutic agents. Cancer Treatment
and Practice, 4th edn., BA Chabner, and DL Longo, Reviews 1982; 9: 3756.
eds., Philadelphia, Lippincott Williams and 18. Munchausen LL and Rahn RO. Biologic and
Wilkins, 2006:332343. chemical effects of cis-dichlorodiammineplatinum
5. Jacobs C, Kalman SM, Tretton M and Weiner MW (II) (NSC-119875) on DNA. Cancer Chemotherapy
Renal handling of cis-diamminedichloroplatinum(II). Reports 1975; 59: 643646.
Cancer Treatment Reports 1980; 64: 12231226. 19. Harder HC, Smith RG and LeRoy AF. Template
6. Choie DD, del Campo AA and Guarino AM. primer inactivation by cis- and trans-
Subcellular localization of cis-dichlorodiamminepl dichlorodiammine platinum for human DNA
atinum(II) in rat kidney and liver. Toxicology and polymerase alpha, beta, and Rauscher murine
Applied Pharmacology 1980; 55: 245252. leukemia virus reverse transcriptase, as a
7. Litterst CL, Gram TE, Dedrick RL, LeRoy AF and mechanism of cytotoxicity. Cancer Research 1976;
Guarino AM. Distribution and disposition of 36: 38213829.
platinum following intravenous administration 20. Plooy AC, van DM and Lohman PH. Induction
of cis-diamminedichloroplatinum(II) (NSC and repair of DNA cross-links in Chinese hamster
119875) to dogs. Cancer Research 1976; 36: 2340 ovary cells treated with various platinum
2344. coordination compounds in relation to platinum
8. Campbell AB, Kalman SM and Jacobs C. Plasma binding to DNA, cytotoxicity, mutagenicity, and
platinum levels: relationship to cisplatin dose and antitumor activity. Cancer Research 1984; 44:
nephrotoxicity. Cancer Treatment Reports 1983; 67: 20432051.
169172. 21. Masuda H, Tanaka T and Takahama U. Cisplatin
9. Cvitkovic E, Spaulding J, Bethune V, Martin J and generates superoxide anion by interaction with
Whitmore WF. Improvement of cis- DNA in a cell-free system. Biochemical Biophysical
dichlorodiammineplatinum (NSC 119875): Research Communications 1994; 203: 11751180.
therapeutic index in an animal model. Cancer 22. Tanaka-Kagawa T, Kitahara J, Seko Y, Toyoda H,
1977; 39: 13571361. Imura N and Naganuma A . Reduced sensitivity
10. Weiner IM. Transport of weak acids and bases. In: of HeLa cells to cis-platinum by simultaneous
Handbook of Physiology. Renal Physiology, J Orloff, overexpression of copper, zinc-superoxide
and RW Berliner, eds., Baltimore, Williams and dismutase and catalase. Biochemistry and
Wilkins, 1973:521554. Pharmacology 1999; 57: 545548.
11. Sarstein R, Miller P and Guttenplan JB. Uptake 23. Godwin AK, Meister A, ODwyer PJ, Huang CS,
and metabolism of cisplatin by rat kidney. Kidney Hamilton TC and Anderson ME. High resistance
International 1984; 25: 753758. to cisplatin in human ovarian cancer cell lines is
12. Litterst CL and Magin R. Alterations in plasma associated with marked increase of glutathione
pharmacokinetics of cisplatin in tumor-bearing rats. synthesis. Proceedings of the National Academy of
Cancer Chemotherapy and Pharmacology 1988; 22: Sciences of the United States of America 1992; 89:
14. 30703074.
13. Rosenberg B. Anticancer activity of cis-dichlorodia- 24. Hosking LK, Whelan RD, Shellard SA, Bedford P
mmineplatinum(II) and some relevant chemistry. and Hill BT. An evaluation of the role of
Cancer Treatment Reports 1979; 63: 14331438. glutathione and its associated enzymes in the

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 13

expression of differential sensitivities to 37. Dentino M, Luit FC, Yum MN, Williams SD and
antitumour agents shown by a range of human Einhom LH. Long term effect of cis-
tumour cell lines. Biochemistry and Pharmacology diamminedichloride platinum (CDDP) on renal
1990; 40: 18331842. function and structure in man. Cancer 1978; 41:
25. Pattanaik A, Bachowski G, Laib J, Lemkuil D, Shaw 12741281.
CF, III, Petering DH, Hitchcock A and Saryan L. 38. Brillet G, Deray G, Jacquiaud C, Mignot L, Bunker
Properties of the reaction of cis-dichlorodiammine D, Meillet D, Raymond F and Jacobs C. Long-term
platinum(II) with metallothionein. Journal of renal effect of cisplatin in man. American Journal
Biological Chemistry 1992; 267: 1612116128. of Nephrology 1994; 14: 8184.
26. Kelley SL, Basu A, Teicher BA, Hacker MP, Hamer 39. Dobyan DC, Levi J, Jacobs C, Kosek J and Werner
DH and Lazo JS. Overexpression of MW. Mechanism of cis-platinum nephrotoxicity:
metallothionein confers resistance to anticancer II. Morphologic observations. Journal of
drugs. Science 1988; 241: 18131815. Pharmacology and Experimental Therapeutics 1980;
27. Borst P, Evers R, Kool M and Wijnholds J. A family 213: 551556.
of drug transporters: the multidrug resistance- 40. Schumer M, Colombel MQ, Sawczuk IS, Gobe G,
associated proteins. Journal of the National Cancer Connor J, OToole KM, Olsson CA, Wise GJ and
Institute 2000; 92: 12951302. Buttyan R. Morphologic, biochemical, and
28. Reed E. Platinum-DNA adduct, nucleotide molecular evidence of apoptosis during the
excision repair and platinum based anti-cancer reperfusion phase after brief periods of renal
chemotherapy. Cancer Treatment Reviews 1998; 24: ischemia. American Journal of Pathology 1992; 140:
331344. 831838.
29. Villella J, Marchetti D, Odunsi K, Rodabaugh K, 41. Townsend DM, Deng M, Zhang L, Lapus MG and
Driscoll DL and Lele S. Response Hanigan MH. Metabolism of cisplatin to a
of combination platinum and gemcitabine nephrotoxin in proximal tubule cells. Journal of the
chemotherapy for recurrent epithelial ovarian American Society of Nephrology 2003; 14: 110.
carcinoma. Gynecologic Oncology 2004; 95: 539545. 42. Sarstein R, Winston J, Moel D, Dikman S and
30. Aebi S, Fink D, Gordon R, Kim HK, Zheng H, Fink Guttenplan J. Cisplatin nephrotoxicity: insights
JL and Howell SB. Resistance to cytotoxic drugs in into mechanism. International Journal of Andrology
DNA mismatch repair-decient cells. Clinical 1987; 10: 325346.
Cancer Research 1997; 3: 17631767. 43. Fraval HN, Rawlings CJ and Roberts JJ. Increased
31. Drummond JT, Anthoney A, Brown R and sensitivity of UV-repair-decient human cells to
Modrich P. Cisplatin and adriamycin resistance are DNA bound platinum products which unlike
associated with MutLalpha and mismatch repair thymine dimers are not recognized by an
deciency in an ovarian tumor cell line. Journal of endonuclease extracted from Micrococcus luteus.
Biological Chemistry 1996; 271: 1964519648. Mutation Research 1978; 51: 121132.
32. Nehme A, Baskaran R, Nebel S, Fink D, Howell SB, 44. Megyesi J, Andrade L, Vieira JM, Jr., Sarstein RL
Wang JY and Christen RD. Induction of JNK and and Price PM.Coordination of the cell cycle is an
c-Abl signalling by cisplatin and oxaliplatin in important determinant of the syndrome of acute
mismatch repair-procient and -decient cells. renal failure. American Journal of Renal Physiology
British Journal of Cancer 1999; 79: 11041110. 2002; 283: F810F816.
33. Lebwohl D and Canetta R. Clinical development 45. Megyesi J, Udvarhelyi N, Sarstein RL and
of platinum complexes in cancer therapy: an Price PM. The p53-independent activation of
historical perspective and an update. European transcription of p21 WAF1/CIP1/SDI1 after
Journal of Cancer 1998; 34: 15221534. acute renal failure. American Journal of
34. Ries F and Klastersky J. Nephrotoxicity induced by Physiology 1996; 271: F1211F1216.
cancer chemotherapy with special emphasis on 46. Megyesi J, Sarstein RL and Price PM. Induction
cisplatin toxicity. American Journal of Kidney of p21WAF1/CIP1/SDI1 in kidney tubule cells
Diseases 1986; 8: 368379. affects the course of cisplatin-induced acute renal
35. Blachley JD and Hill JB. Renal and electrolyte failure. Journal of Clinical Investigation 1998; 101:
disturbances associated with cisplatin. Annals of 777782.
Internal Medicine 1981; 95: 628632. 47. Price PM, Sarstein RL and Megyesi J. Protection
36. Madias NE and Harrington JT. Platinum of renal cells from cisplatin toxicity by cell cycle
nephrotoxicity. American Journal of Medicine 1978; inhibitors. American Journal of Renal Physiology
65: 307314. 2004; 286: F378F384.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
14 K. Barabas et al.

48. Clarkson TW. The pharmacology of mercury 60. Hanigan MH, Lykissa ED, Townsend DM, Ou CN,
compounds. Annual Review of Pharmacology 1972; Barrios R and Lieberman MW. Gamma-glutamyl
12: 375406. transpeptidase-decient mice are resistant to the
49. Levi J, Jacobs C, Kalman SM, McTigue M, Weiner nephrotoxic effects of cisplatin. American Journal
MW. Mechanism of cis-platinum nephrotoxicity: of Pathology 2001; 159: 18891894.
I. Effects of sulfhydryl groups in rat kidneys. 61. Hanigan MH, Gallagher BC, Taylor PT, Jr. and
Journal of Pharmacology and Experimental Large MK. Inhibition of gamma-glutamyl
Therapeutics 1980; 213: 545550. transpeptidase activity by acivicin in vivo protects
50. Lieberthal W, Triaca V and Levine J. Mechanisms the kidney from cisplatin-induced toxicity. Cancer
of death induced by cisplatin in proximal tubular Research 1994; 54: 59255929.
epithelial cells: apoptosis vs. necrosis. American 62. Bemareggi A, Torti L, Faeino RM, Carini M, Depta
Journal of Physiology 1996; 270: F700F708. G, Casetta B, Farrell N, Spadacini S, Ceserani R
51. Kroemer G and Reed JC. Mitochondrial control of and Tognella S. Characterization of cisplatin-
cell death. Nature Medicine 2000; 6: 513519. glutathione adducts by liquid chromatography-
52. Brady HR, Kone BC, Stromski ME, Zeidel ML, mass spectrometry. Evidence for their formation
Giebisch G and Gullans SR. Mitochondrial injury: in vitro but not in vivo after concomitant
an early event in cisplatin toxicity to renal administration of cisplatin and glutathione to rats
proximal tubules. American Journal of Physiology and cancer patients. Journal of Chromatography B
1990; 258: F1181F1187. Biomedical Applications 1995; 669: 247263.
53. Kruidering M, van de WB, de HE, Mulder GJ and 63. Ishikawa T and Ii-Osman F. Glutathione-
Nagelkerke JF, Cisplatin-induced nephrotoxicity associated cis-diamminedichloroplatinum(II)
in porcine proximal tubular cells: mitochondrial metabolism and ATP-dependent efux from
dysfunction by inhibition of complexes I to IV of leukemia cells. Molecular characterization of
the respiratory chain. Journal of Pharmacology and glutathione-platinum complex and its biological
Experimental Therapeutics 1997; 280: 638649. signicance. Journal of Biological Chemistry 1993;
54. Davis CA, Nick HS and Agarwal A. Manganese 268: 2011620125.
superoxide dismutase attenuates cisplatin-induced 64. Gosland M, Lum B, Sehimmelpfennig J, Baker J
renal injury: importance of superoxide. Journal of the and Doukas M. Insights into mechanisms of
American Society of Nephrology 2001; 12: 26832690. cisplatin resistance and potential for its clinical
55. Lau AH. Apoptosis induced by cisplatin reversal. Pharmacotherapy 1996; 16: 1639.
nephrotoxic injury. Kidney International 1999; 56: 65. Hanigan MH, Gallagher BC, Townsend DM and
12951298. Gabarra V. Gamma-glutamyl transpeptidase
56. Ye J, Wang S, Leonard SS, Sun Y, Butterworth L, accelerates tumor growth and increases the
Antonini J, Ding M, Rojanasakul Y, Vallyathan V, resistance of tumors to cisplatin in vivo.
Castranova V and Shi X. Role of reactive oxygen Carcinogenesis 1999; 20: 553559.
species and p53 in chromium(VI)-induced 66. Arany I and Sarstein RL. Cisplatin
apoptosis. Journal of Biological Chemistry 1999; nephrotoxicity. Seminars in Nephrology 2003; 23:
274: 3497434980. 460464.
57. Cummings BS and Schnellmann RG. Cisplatin- 67. Sarstein R, Miller P, Dikman S, Lyman N and
induced renal cell apoptosis: caspase 3-dependent Shapiro C. Cisplatin nephrotoxicity in rats: defect
and -independent pathways. Journal of in papillary hypertonicity. American Journal of
Pharmacology and Experimental Therapeutics Physiology 1981; 241: F175F185.
2002; 302: 817. 68. Chopra S, Kaunan JS, Jones TW, Hong WK, Gehr
58. Xiao T, Choudhary S, Zhang W, Ansari NH and MK, Hamburger RJ, Flaraenbaum W and Trump
Salahudeen A. Possible involvement of oxidative BF. Cis-diamminedichlorplatinum-induced acute
stress in cisplatin-induced apoptosis in LLC-PK1 renal failure in the rat. Kidney International 1982;
cells. Journal of Toxicology and Environmental 21: 5464.
Health A 2003; 66: 469479. 69. Schor N, Ichikawa I, Rennke HG, Troy JL and
59. Townsend DM and Hanigan MH. Inhibition of Brenner BM. Pathophysiology of altered
gamma-glutamyl transpeptidase or cysteine glomerular function in aminoglycoside-treated
S-conjugate beta-lyase activity blocks the rats. Kidney International 1981; 19: 288296.
nephrotoxicity of cisplatin in mice. Journal of 70. Daugaard G and Abildgaard U. Cisplatin
Pharmacology and Experimental Therapeutics 2002; nephrotoxicity. A review. Cancer Chemotherapy
300: 142148. and Pharmacology 1989; 25: 19.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 15

71. Ogilvie GK, Krawiec DR, Gelberg HB, Twardock diamminedichloro-platinum(II)-induced


AR, Reschke RW and Richardson BC. Evaluation cytotoxicity, DNA cross-linking, and gamma-
of a short-term saline diuresis protocol for the glutamyl transpeptidase inhibition. Cancer
administration of cisplatin. American Journal of Research 1986; 46: 27452750.
Veterinary Research 1988; 49: 10761078. 82. Beny JM, Jacobs C, Sikic B, Halsey J and Borch RF.
72. Ogilvie GK, Straw RC, Jameson VJ, Walters LM, Modication of cisplatin toxicity with
Lafferty M, Powers BE, Henkel SE and Withrow SJ. diethyldithiocarbamate. Journal of Clinical
Prevalence of nephrotoxicosis associated with a Oncology 1990; 8: 15851590.
four-hour saline solution diuresis protocol for 83. Jacobs C, Kaubisch S, Halsey J, Lurn BL, Gosland
the administration of cisplatin to dogs with M, Coleman CN and Sikic BT. The use of
naturally developing neoplasms. Journal of the probenecid as a chemoprotector against cisplatin
American Veterinary Medical Association 1993; nephrotoxicity. Cancer 1991; 67: 15181524.
202: 18451848. 84. Wu YJ, Muldoon LL and Neuwelt EA. The
73. Ostrow S, Egorin MJ, Hahn D, Markus S, Aisner J, chemoprotective agent N-acetylcysteine blocks
Chang P, LeRoy A, Bachur NR and Wiernik PH. cisplatin-induced apoptosis through caspase
High-dose cisplatin therapy using mannitol versus signaling pathway. Journal of Pharmacology and
furosemide diuresis: comparative Experimental Therapeutics 2005; 312: 424431.
pharmacokinetics and toxicity. Cancer Treatment 85. Howell SB, Pfeie CE, Wung WE and Olshen RA.
Reports 1981; 65: 7378. Intraperitoneal cis-diamminedichloroplatinum
74. Pera MF Jr, Zook BC and Harder HC. Effects of with systemic thiosulfate protection. Cancer
mannitol or furosemide diuresis on the Research 1983; 43: 14261431.
nephrotoxicity and physiological disposition of 86. Dickey DT, Wu YJ, Muldoon LL and Neuwelt EA.
cis-dichlorodiammineplatinum-(II) in rats. Cancer Protection against cisplatin-induced toxicities by
Research 1979; 39: 12691278. N-acetylcysteine and sodium thiosulfate as assessed
75. Al-Sarraf M. Clinical trial of cis-platinum (NSC- at the molecular, cellular, and in vivo levels.
119875): hydration with and without mannitol in Journal of Pharmacology and Experimental
patients with previously treated advanced Therapeutics 2005; 314: 10521058.
malignant melanoma. Proc Am Assoc Cancer Res 87. Esposito M, Viale M, Vannozzi MO, Zicca A,
and ASCO 1979; 20: 185. Cadoni A, Merlo F and Gogioso L. Effect of the
76. Belt RJ and Taylor S. Randomized trial of hydration antiarrhythmic drug procainamide on the toxicity
and mannitol in preventing cis-platinum induced and antitumor activity of cis-diamminedic-
nephrotoxicity. Proceedings of the American hloroplatinum(II). Toxicology and Applied
Association for Cancer Research and the American Pharmacology 1996; 140: 370377.
Society of Clinical Oncology 1979; 20: 388. 88. Viale M, Vannozzi MO, Pastrone I, Mariggio MA,
77. Litterst CL. Alterations in the toxicity of cis- Zicca A, Cadoni A, Cafaggi S, Tolino G, Lunardi G,
dichlorodiammineplatinum-II and in tissue Civalleri D, Lindup WE and Esposito M.
localization of platinum as a function of NaCl Reduction of cisplatin nephrotoxicity by
concentration in the vehicle of administration. procainamide: does the formation of a cisplatin-
Toxicology and Applied Pharmacology 1981; 61: procainamide complex play a role? Journal of
99108. Pharmacology and Experimental Therapeutics 2000;
78. Yuhas JM and Culo F. Selective inhibition of the 293: 829836.
nephrotoxicity of cis-dichlorodiammineplatinum 89. Vail DM, Elfarrn AA, Cooley AJ, Panciera DL,
(II) by WR-2721 without altering its antitumor MacEwen EG and SoergeJ SA. Methimazole as a
properties. Cancer Treatment Reports 1980; 64: protectant against cisplatin-induced
5764. nephrotoxicity using the dog as a model. Cancer
79. Weiner MW and Jacobs C. Mechanism of cisplatin Chemotherapy and Pharmacology 1993; 33: 2530.
nephrotoxicity. Federation Proceedings 1983; 42: 90. Thamm DH and Vail DM. Preclinical evaluation
29742978. of a sterically stabilized liposome-encapsulated
80. Jones MM, Basinger MA and Holscher MA. Control cisplatin in clinically normal cats. American
of the nephrotoxicity of cisplatin by clinically used Journal of Veterinary Research 1998; 59: 286289.
sulfur-containing compounds. Fundamental and 91. Fox LE, Toshach K, Calderwood-Mays M, Khokhar
Applied Toxicology 1992; 18: 181188. AR, Kubilis P, Perez-Soler R and MacEwen EG.
81. Bodenner DL, Dedon PC, Keng PC and Borch RF. Evaluation of toxicosis of liposome-encapsulated cis-
Effect of diethyldithiocarbamate on cis- bis-neodecanoato-trans-R,R-1,2-diaminocyclohexane

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
16 K. Barabas et al.

platinum (II) in clinically normal cats. American 101. Tayem Y, Johnson TR, Mann BE, Green CJ and
Journal of Veterinary Research 1999; 60: 257263. Motteriini R. Protection against cisplatin-induced
92. Marr AK, Kurzman ID and Vail DM. Preclinical nephrotoxicity by a carbon monoxide-releasing
evaluation of a liposome-encapsulated formulation molecule. American Journal of Physiology. Renal
of cisplatin in clinically normal dogs. American Physiology 2006; 290: F789F794.
Journal of Veterinary Research 2004; 65: 102. Barabas K, Milner R, Farese J, Adin C and Croker
14741478. B. Hyperbilirubinemia protects against cisplatin
93. McGinness JE, Proctor PH, Demopoulos HB, nephrotoxicity. Proceeding of the Veterinary Cancer
Hokanson JA and Kirkpatrick DS. Amelioration of Society 2006; 36.
cis-platinum nephrotoxicity by orgotein 103. Schilsky RL and Anderson T. Hypomagnesemia
(superoxide dismutase). Physiological Chemistry and renal magnesium wasting in patients receiving
and Physics 1978; 10: 267277. cisplatin. Annals of Internal Medicine 1979; 90:
94. Clark JE, Foresti R, Sarathchandra P, Kaur H, 929931.
Green CJ and Motterlim R. Heme oxygenase- 104. Mavichak V, Wong NL, Quamme GA, Magil AB,
1-derived bilirubin ameliorates postischemic Sutton RA and Dirks JH. Studies on the
myocardial dysfunction. American Journal of pathogenesis of cisplatin-induced
Physiology Heart and Circulatory Physiology 2000; hypomagnesemia in rats. Kidney International
278: H643H651. 1985; 28: 914921.
95. Foresti R, Green CJ and Motterlini R. Generation 105. Suh SM, Tashjian AH Jr, Matsuo N, Parkinson DK
of bile pigments by haem oxygenase: a rened and Fraser D. Pathogenesis of hypocalcemia in
cellular strategy in response to stressful insults. primary hypomagnesemia: normal end-organ
Biochemical Society Symposium 2004; 71: 177192. responsiveness to parathyroid hormone, impaired
96. Motterlini R, Gonzales A, Foresti R, Clark JE, parathyroid gland function. Journal of Clinical
Green CJ and Winslow RM. Heme oxygenase-1- Investigation 1973; 52: 153160.
derived carbon monoxide contributes to the 106. Connor TB, Toskes P, MahafFey J, Martin LG,
suppression of acute hypertensive responses Williams JB and Walser M. Parathyroid function
in vivo. Circulation Research 1998; 83: 568577. during chronic magnesium deciency. The Johns
97. Sammut A, Foresti R, Clark JE, Exon DJ, Vesely Hopkins Medical Journal 1972; 131: 100117.
MJ, Sarathchandra P, Green CJ and Motteriini R. 107. Moore AS, Rand WM, Berg J, LHeureux DA and
Carbon monoxide is a major contributor to the Dennis RA. Evaluation of butorphanol and
regulation of vascular tone in aortas expressing cyproheptadine for prevention of cisplatin-
high levels of haeme oxygenase-1. British Journal of induced vomiting in dogs. Journal of the
Pharmacology 1998; 125: 14371444. American Veterinary Medical Association 1994;
98. Otterbein LE, Bach FH, Alam J, Soares M, Tao LH, 205: 441443.
Wysk M, Davis RJ, Flavell RA and Choi AM. 108. Plumb D. In: Veterinary Drug Handbook, 5th edn.,
Carbon monoxide has anti-inammatory effects D Plumb ed., Ames, Blackwell Publishing, 2005:
involving the mitogen-activated protein kinase 103, 162, 275, 520, 573.
pathway. Nature Medicine 2000; 6: 422428. 109. Vail DM, Rodabaugh HS, Conder GA, Boucher JF
99. Lavitrano M, Smolenski RT, Musumeci A, and Mathur S. Efcacy of injectable maropitant
Maceherini M, Slominska E, Di FE, Bracco A, (CereniaTM) in a randomized clinical trial for
Mancini A, Stassi G, Patti M, Giovannoni R, Froio prevention and treatment of cisplatin induced
A, Simeone F, Fomi M, Bacci ML, DAlise G, Cozzi emesis in dogs presented as veterinary patients.
E, Otterbein LE, Yacoub MH, Bach FH and Calise Veterinary and Comparative Oncology 2007; 5: 3846.
F. Carbon monoxide improves cardiac energetics 110. Ellerby RA, Davis HL Jr, Anseld FJ and Ramirez G.
and safeguards the heart during reperfusion after Phase I clinical trial of combined therapy with
cardiopulmonary bypass in pigs. Federation of 5-FU (NSC 19893) and CIS-platinum (II)
American Societies for Experimental Biology Journal diaminedichloride (NSC 119875). Cancer 1974;
2004; 18: 10931095. 34: 10051010.
100. Shiraishi F, Curtis LM, Truong L, Poss K, Visner 111. Green D. Pure tone air conduction threshold. In:
GA, Madsen K, Nick HS and Agarwal A. Heme Handbook of Clinical Audiology, Katz J, ed.,
oxygenase-1 gene ablation or expression Baltimore, Williams and Wilkins, 1972: 6.
modulates cisplatin-induced renal tubular 112. Helson L, Okonkwo E, Anton L and Cvitkovic E.
apoptosis. American Journal of Renal Physiology cis-Platinum ototoxicity. Clinical Toxicology 1978;
2000; 278: F726F736. 13: 469478.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
Review of cisplatin 17

113. Schell MJ, McHaney VA, Green AA, Kun LE, 124. Chun R, Garrett LD, Henry C, Wall M, Smith A
Hayes FA, Horowitz M and Meyer WH. Hearing and Azene NM. Toxicity and efcacy of cisplatin
loss in children and young adults receiving and doxorubicin combination chemotherapy for
cisplatin with or without prior cranial irradiation. the treatment of canine osteosarcoma. Journal of
Journal of Clinical Oncology 1989; 7: 754760. the American Animal Hospital Association 2005; 41:
114. Pollera CF, Marolla P, Nardi M, Ameglio F, Cozzo L 382387.
and Bevere F. Very high-dose cisplatin-induced 125. Berg J, Gebhardt MC and Rand WM. Effect of
ototoxicity: a preliminary report on early and timing of postoperative chemotherapy on survival
long-term effects. Cancer Chemotherapy and of dogs with osteosarcoma. Cancer 1997; 79:
Pharmacology 1988; 21: 6164. 13431350.
115. Pickrell JA, Oehme FW and Cash WC. Ototoxicity 126. Knapp DW, Richardson RC, Bonney PL and
in dogs and cats. Seminars in Veterinary Medicine Hahn K. Cisplatin therapy in 41 dogs with
and Surgery (Small Animal) 1993; 8: 4249. malignant tumors. Journal of Veterinary Internal
116. Ozols RF and Young RC. High-dose cisplatin Medicine 1988; 2: 4146.
therapy in ovarian cancer. Seminars in Oncology 127. Vail DM, Kurzman ID, Glawe PC, OBrien MG,
1985; 12(Suppl. 6): 2130. Chun R, Garrett LD, Obradovich JE, Fred RM, III,
117. Sorensen JB, Andersen MK and Hansen HH. Khanna C, Colbem GT and Working PK.
Syndrome of inappropriate secretion of STEALTH liposome-encapsulated cisplatin (SPI-
antidiuretic hormone (SIADH) in malignant 77) versus carboplatin as adjuvant therapy for
disease. Journal of International Medicine 1995; spontaneously arising osteosarcoma (OSA) in the
238: 97110. dog: a randomized multicenter clinical trial. Cancer
118. Shapiro W, Fossum TW, Kitchell BE, Couto CG Chemotherapy and Pharmacology 2002; 50: 131136.
and Theilen GH. Use of cisplatin for treatment of 128. Withrow SJ, Liptak JM, Straw RC, Demell WS,
appendicular osteosarcoma in dogs. Journal of the Jameson VJ, Powers BE, Johnson JL, Brekke JH
American Veterinary Medical Association 1988; 192: and Douple EB. Biodegradable cisplatin polymer
507511. in limb-sparing surgery for canine osteosarcoma.
119. Rraegel SA, Madewell BR, Simonson E and Annals of Surgical Oncology 2004; 11: 705713.
Gregory CR. Osteogenic sarcoma and cisplatin 129. Hahn KA, Richardson RC, Blevins WE, Lenz SD
chemotherapy in dogs: 16 cases (19861989). and Knapp DW. Intramedullary cisplatin
Journal of the American Veterinary Medical chemotherapy: experience in four dogs with
Association 1991; 199: 10571059. osteosarcoma. Journal of Small Animal Practice
120. Straw RC, Withrow SJ, Richter SL, Powers BE, Klein 1996; 37: 187192.
MK, Postorino NC, LaRue SM, Ogilvie GK, Vail 130. Shapiro W, Kitchell BE, Fossum TW, Couto CG
DM and Morrison WB. Amputation and cisplatin and Theilen G. Cisplatin for treatment of
for treatment of canine osteosarcoma. Journal of transitional cell and squamous cell carcinomas in
Veterinary Internal Medicine 1991; 5: 205210. dogs. Journal of the American Veterinary Medical
121. Thompson JP and Fugent MJ. Evaluation of Association 1988; 193: 15301533.
survival times after limb amputation, with and 131. Dewit L. Combined treatment of radiation and
without subsequent administration of cisplatin, cisdiamminedichloroplatinum (II): a review of
for treatment of appendicular osteosarcoma in experimental and clinical data. International
dogs: 30 cases (19791990). Journal of the Journal of Radiation Oncology Biology Physics 1987;
American Veterinary Medical Association 1992; 13: 403426.
200: 531533. 132. Begg AC. Cisplatin and radiation: interaction
122. Berg J, Weinstein MJ, Schelling SH and Rand WM. probabilities and therapeutic possibilities.
Treatment of dogs with osteosarcoma by International Journal of Radiation Oncology Biology
administration of cisplatin after amputation or Physics 1990; 19: 11831189.
limb-sparing surgery: 22 cases (19871990). 133. Coughlin CT and Richmond RC. Biologic and
Journal of the American Veterinary Medical clinical developments of cisplatin combined with
Association 1992; 200: 20052008. radiation: concepts, utility, projections for new
123. Mauldin GN, Matus RE, Withrow SJ and Patnaik trials, and the emergence of carboplatin. Seminars
AK. Canine osteosarcoma. Treatment by amputation in Oncology 1989; 16(Suppl. 6): 3143.
versus amputation and adjuvant chemotherapy 134. Douple EB. Platinum-radiation interactions.
using doxorubicin and cisplatin. Journal of National Cancer Institute Monographs 1988; 6:
Veterinary Internal Medicine 1988; 2: 177180. 315319.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118
18 K. Barabas et al.

135. Al-Sarraf M, LeBlanc M, Giri PG, Fu KK, Cooper Calderwood-Mays M and Kurzman ID. Use of
J, Vuong T, Forastiere AA, Adams G, Sakr WA, cis-bis-neodecanoato-trans-R,R-1,2-
Sehuller DE and Ensley JF. Chemoradiotherapy diaminocyclohexane platinum (II), a liposomal
versus radiotherapy in patients with advanced cisplatin analogue, in cats with oral squamous cell
nasopharyngeal cancer: phase III randomized carcinoma. American Journal of Veterinary
Intergroup study 0099. Journal of Clinical Oncology Research 2000; 61: 791795.
1998; 16: 13101317. 147. Theon AP, Pascoe JR, Carlson GP and Krag DN.
136. Nadeau ME, Kitchell BE, Rooks RL and LaRue Intratumoral chemotherapy with cisplatin in
SM. Cobalt radiation with or without low-dose oily emulsion in horses. Journal of the American
cisplatin for treatment of canine naso-sinus Veterinary Medical Association 1993; 202:
carcinomas. Veterinary Radiology & Ultrasound 261267.
2004; 45: 362367. 148. Theon AP, Pascoe JR, Madigan JE, Carlson G and
137. Lana SE, Dernell WS, Lafferty MH, Withrow SJ Metzger L. Comparison of intratumoral
and LaRue SM. Use of radiation and a slow-release administration of cisplatin versus bleomycin for
cisplatin formulation for treatment of canine nasal treatment of periocular squamous cell carcinomas
tumors. Veterinary Radiology & Ultrasound 2004; in horses. American Journal of Veterinary Research
45: 577581. 1997; 58: 431436.
138. Freeman KP, Hahn KA, Harris FD and King GK. 149. Theon AP, Pascoe JR and Meagher DM.
Treatment of dogs with oral melanoma by Perioperative intratumoral administration of
hypofractionated radiation therapy and platinum- cisplatin for treatment of cutaneous tumors in
based chemotherapy (19871997). Journal of equidae. Journal of the American Veterinary
Veterinary Internal Medicine 2003; 17: 96101. Medical Association 1994; 205: 11701176.
139. Kitchell BE, Brown DM, Luck EE, Woods LL, 150. Theon AP , Pascoe JR , Galuppo LD , Fisher PE ,
Orenberg EK and Bloch DA. Intralesional implant Griffey SM and Madigan JE . Comparison of
for treatment of primary oral malignant melanoma perioperative versus postoperative intratumoral
in dogs. Journal of the American Veterinary Medical administration of cisplatin for treatment of
Association 1994; 204: 229236. cutaneous sarcoids and squamous cell
140. Moore AS, Kirk C and Cardona A. Intracavitary carcinomas in horses. Journal of the American
cisplatin chemotherapy experience with six dogs. Veterinary Medical Association 1999; 215:
Journal of Veterinary Internal Medicine 1991; 5: 16551660.
227231. 151. Bergman PJ, MacEwen EG, Kurzman ID, Henry
141. Myers CE and Collins JM. Pharmacology of CJ, Hammer AS, Knapp DW, Hale A, Kruth SA,
intraperitoneal chemotherapy. Cancer Investigation Klein MK, Klausner J, Norris AM, McCaw D,
1983; 1: 395407. Straw RC and Withrow SJ. Amputation and
142. Markman M. Intraperitoneal antineoplastic carboplatin for treatment of dogs with
agents for tumors principally conned to the osteosarcoma: 48 cases (19911993). Journal of
peritoneal cavity. Cancer Treatment Review 1986; Veterinary Internal Medicine 1996; 10: 7681.
13: 219242. 152. Kent MS, Strom A, London CA and Seguin B.
143. Charney SC, Bergman PJ, McKnight JA, Farrelly J, Alternating carboplatin and doxorubicin as
Novosad CA, Leibman NF and Camps-Palau MA. adjunctive chemotherapy to amputation or limb-
Evaluation of intracavitary mitoxantrone and sparing surgery in the treatment of appendicular
carboplatin for treatment of carcinomatosis, osteosarcoma in dogs. Journal of Veterinary
sarcomatosis, and mesothelioma, with or without Internal Medicine 2004; 18: 540544.
malignant effusions: a retrospective analysis of 12 153. Chun R, Knapp DW, Widmer WR, DeNicola DB,
cases (19972002). Veterinary and Comparative Glickman NW, Kuczek T, Degortari A and Han
Oncology 2005; 3: 171181. CM. Phase II clinical trial of carboplatin in canine
144. Knapp DW, Richardson RC, DeNicola DB, Long GG transitional cell carcinoma of the urinary bladder.
and Blevins WE. Cisplatin toxicity in cats. Journal of Journal of Veterinary Internal Medicine 1997; 11:
Veterinary Internal Medicine 1987; 1: 2935. 279283.
145. Shapiro W. Cisplatin chemotherapy. In: Current 154. Kirpensteijn J, Teske E, Kik M, Klenner T and
Veterinary Therapy X, Kirk RW, ed., Philadelphia, Rutteman GR. Lobaplatin as an adjuvant
WB Saunders Co, 1989: 497501. chemotherapy to surgery in canine appendicular
146. Fox LE, Rosenthal RC, King RR, Levine PB, Vail osteosarcoma: a phase II evaluation. Anticancer
DM, Helfand SC, MacEwen EG, Perez-Soler R, Research 2002; 22: 27652770.

2008 The Authors. Journal compilation 2008 Blackwell Publishing Ltd, Veterinary and Comparative Oncology, 6, 1, 118

Das könnte Ihnen auch gefallen