+ All Categories
Home > Documents > 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

Date post: 06-Apr-2018
Category:
Upload: kannan-narayanan
View: 216 times
Download: 0 times
Share this document with a friend

of 12

Transcript
  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    1/12

    ~ 9 - - 7 / 4 d c7 */>/- r r r , / > . w ~ ~ c ~ ~ p h o ~ /WZ~-?JL.&5 h p 1 p ~ A u . -PW * j/lF y , ~ ~ A ~S F M I +c - MMM C OW / G ~ v o i J c c I/1- f l M R

    m r m c y A ~ N p C Z F - m c A t M C OZ i z - L ' p ~ ~c~10.cI $ i F - /su=Nephrotoxicity Induced b y Cancer Chemotherapy WithSpecial Emphasis on Cisplatin Toxicity

    F. Ries. MD, and J. Klastersky, MDRena liailure in cancer patients Is a common problem in oncology; this complicatlon is frequently multlfactorial

    In origih. Several antineoplastlc agents are potentially nephrotoxlc; previous renal mpairment as well aa comblnr.tlons with other nephrotoxic drugs may Increase he risk of nephrotoxiclty during administration of chemotherapy.Methotrexate-related renal damage most frequently occula with hlghdose therapy and can be av o ld d by t o malkaline diuresis and administration of fo llnlc acid. Renal dysfunction secondary to semustlne (CHACNU) 18clearly related o cumulative doses in excess to 1,200 mglma; the onset may be delayed and renal failure pro greudespite drug discontinuation. Streptozotocin is also nephrotoxic and may cause proteinuria and renal tubularacidosis; progressive renal failu-dicted by a close monitoring of protelnuria and preventd by drugdiscontinuance. Mltomycin-associated enal failure frequently presents with sign8 of microanglopathlc hemolyticanemia; renal failure is usually delayed but occasionally, It may be rapidly progressive despite drug dlscontlnu.ance. Clsplatin nephrotoxicity is clearly dose-related and used to be considered dose llmltlng. Renal In su ff lc le n~ ~can be prevented by hydration and forced dlsuresis; i n addltlon, hyperhydratlon with mannltol-Induced 8allnedluresis may allow administration of high doses and thus circumvent the dose-limiting effect of clsplatln-Inducedrenal toxicity. Cisplatin-induced renal magnesium wasting occurs frequently and should be supplemented. C?!-;.rapproaches to reduce c~splatin ephrotoxicity are currently under investigation and are diacusOed.9 1986 by the National Kldney Foundaton, Inc.INDEX WORDS: Nephrotoxic drugs; chemotherapy; renal failure; cisplatln; methotrexate; mltomycin; semustine;streptozotocin; cancer.

    S NCE THE KIDNEY is highly susceptible totoxic injury by a multitude of different drugs.it is not surprising that several antineoplasticagents may exert potent nephrotoxicity. In cancerpatients. however, one must always be aware ofadditive or synergistic efiects that may potentiatechemotherapy-induced nephrotoxicity. Cancer pa-tients are frequently treated with nephrotoxic anti-biotics (eg, aminoglycosides or amphotericin B).They may suffer from radiation-induced nephro-toxicity. or present with hypercalcemia. hyperuri-cemia. lysozymuria. cancer-related microan-giopathic hemolysis. or disseminated intravascularcoagulopathy. These patients also may presentwith immune complex-mediated glomerulopathyor paraprotein-'felated nephropathy, and with amy-loidosis as well as minimal-change nephritis. They

    contrast media may be potentially harmful to thekidney; cancer patients frequently suffer fromurinary tract infections. It is. therefore, importantto take into account every potentially harmful ef-fect when looking for chemotherapy-relatednephrotoxicity.The agents whose potential for nephrotoxicityhas been clearly established will be discussed i::the following review. These are methotrexate. :..platin. semustine (methyl-CCNU), streptozotocrn.mithramycin. mitomycin C, and azacytidine. Cis-platin nephrotoxicity will be reviewed in greaterdetail. In addition. the reader is informed that theproblem of chemotherapy-related nephrotoxicityhas been analyzed in several review articles.'.'METHOTREXATEmay have direct infiltration of the kidney by tumdr Methotrexate (MTX), an inhibitor of folate syn-cells as well as an obstructive nephropathy caused thesis. is one of the earliest and one of the mostby lower urinary tract invasion and compression. used agents in cancer chemotherapy. Used forDifferent radiologic investigations with iodine more than 30 years. MTX has shown activity in

    From rhe Senic e de Mid edn e lnrtrnr er Laboraroire d' ln - great number of tumors. such as leukemia. !I '\.esrigarion Clinique H. 7hgnon. Cenrre des Tumeurs de phoma. choriocarcinoma, osteosarcoma. andI'Universire Ljbre de Bturelles. lr~srirurJules Border. B n u - mors of the breast, h e d and neck, and lung."elles.

    Address reprinr rcquesrs ro F: Ries. MD. mrirur Jules Bor- Since MTX is excreted primarily by the kidney.der. I Rue Heger- order. IWO Bnuelles. Belgique. any changes in renal function will have an effectC 198 6 bv rhe National Ki dt~ e; ou&iion. Inc. on hfTX plasma levels arid MTX clearance, with02 n-6386/86/08os-oo14soj. 0010 subsequent exposuro of the whole organism to per-

    368 American Jourri6l of IOdn6y 3iseases. Vol Vlli. No 5 (N ovember ) . 1986: pp 368-379

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    2/12

    EPHROTOXICITY OF CYTOSTATICSy elevated c on cen tr atio ns of t he d r ~ g . ~ . ~ . ~liferating or cycling cells. since those of the

    e marrow and gastrointestinal tract are mostusceptible to the toxic effects of M TX and intox-with MTX secondary to renal failure, pri-

    result in marrow hypop laiia and sev ere mu-cositis. Apart from causing important systemictoxicity, high plasma concentrations of MTX arealso nephrotoxic per se, and renal failur e has beenimplicated in 20% of the deaths associated withh ighdose MTX admin i s t ra t i~ n .~

    qeveral mechanisms may contribute to MTX-:r~ ate d enal failure: p recipitation of M TX inrenal tubules, direct antimitotic effect on renaltubular cells, and possibly, alteration of glomeru-Iar filtration rate (GFR).Among these mecha-nisms, tubular precipitation pro ducing obstructiverenal failure is the best know n and the most widelyaccepted. MTX solubility has been shown tochange with urinary pH, and the drug was shownto be two to ten times more soluble at pH 6.9 thanat pH 5.7.9.'0 Precipitation o f the dr ug may occurin concentrated and acid urine resulting in renal~ b u l a rbstruction by an amorphous yellow mate-rial; this has been shown at autopsy examinationsof MTX-treated patients dying from acute renalf a i l ~ r e . ~he amorphous material was identified asMTX by immunofluorescent techniques. The riskof tubular MTX precipitation can be minimized by< hydration as well as alkalinization. I - ' ) Since hy-

    : dration and alkalinization programs have been in-troduced into clinical prac tice, the incidence of re-nal toxicity has decreased dramatically.3th er observations suggest that MT X might in-

    2ce renal failure without tubu lar precipitation be-cause oliguria or anuria are rarely found. and trueobstructive nephropathy has only been reported ina few case repc ~r ts . ~ince MTX reaches high con-centrations for prolonged periods in h e urine, re-nal tubular epi thelialc ells undergoing renewal orrepair could be potentially susceptible to this con-

    - centration by time effect of the drug. 4A high-dose MTX-related decrease in GFR wasobserved in pediatric and adult pa tients,I5.l6 butthe pathogenetic mecha nism of this phenomeno n isstill unclear.MTX nephrotoxicity may be predicted by

    , several tests: ( 1 ) ~ o w ' u r i n a r ~H during high-doseMTX infusion may be associated with impendingtoxicity. In high-dose MTX programs. urinary pHshould be maintained above pH 7 by intravenous

    (IV) or o ra l b ica rbona te ad mi n i~ t ra t i o n .~2) A riseof serum creatinine value from baseline pretreat-ment value may identify patients at risk for addi-tional renal damage. For these patients, adminis-tration of leucovorin rescue as well as vigoroushydration and alkalinization should be consid-ered.9 (3) Monitoring of serum MTX levels is ofprimordial importance and identifies patientsneeding additional rescue measures. Leucovorinrescue should be provided until M TX s erum levels -,fal l below lo- ' m o l / ~ .7-19 i - ' i- i P '-/dl z 6 . 0 9 )

    In patients with established nephrotoxicity,leucovorin rescue treatment may counteract thecytotoxic effects of MTX. Leucovorin (tetrahy-drofolate), inhibitins MTX activity by competi-tion. is administered in high-dose MTX programsat conventional dose s of 15 mg, r epeated at regularintervals12; since MTX concentrations are some-times extremely high in MTX-induced renalfailure, the doses of leucovorin rescue should beapprop r i a t e ly i nc rea~ ed .~owering of M TX levelsby means of peritoneal dialysis, hemodialysis. orhemoperfusion s eem s to be of marginal v a l ~ e . ~ ~ - ~ ~Since, in most cases. patients are not oliguric oranuric in MTX-related renal failure, high fluid in-take with alkalinization should always be main-tained. Apart from v igorous hyd ration, alkaliniza-tion. and administration of leucovorin duringhigh-dose MTX therapy, care should be taken toavoid the drug in patients who suffer from pre-liminary renal insufficiency or to postpone its usein patients who are dehydrated; urine acidifyingagents and salicglate derivatives should be avoidedbefore and during MTX treatment. Good knowl-edge and careful application of these efficientmeasures for prevention of MTX-related nephro-toxicity have been associated with the near disap-pearance of this potentially harmful complication.

    SEMUSTINE (METHYL-CCNU)Semustine. l ike the other nitrosourea com-

    pounds. is a highly lipid-soluble drug that hassome clinical activity in neoplasms of the brain.gastrointestinal tract. as well as malignant mela-noma. Nephrotoxicity of this compound had beendescribed in early preclinical toxicology experi-m e n t ~ , ~ , ' , ~ 'ut evidence for possible nephrotoxic-ity in humans appeared only much later in 1978and 1979.26.27 ince then. numerou s othe r reportso n s e m u s t i n e n e p h r o t o x i c i r y h a v e b e e np u b l i s l ~ e d . ~ ~ - ~ ~

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    3/12

    RlES AND KLASTERSKY

    The most frequent clinical manifestation is aslowly rising serum creatinine level with progres-sive irreversible renal failure. Pathologic findingsare generally consistent with the presence of sm alland atrophic kidneys, signs of tubular atroph y, hy-alinization, glomerulosclerosis, and interstitialfibrosis.32Semustine nephrotoxicity is cumulative; renaldysfunction has been noted in 26%of patients whoreceived total cumulative do ses in excess of 1,400m g / n ~ ~ . ~ I median cumulative dose at whichnephrotoxicity is likely to occur has been esti-mated to be near 2,000 mg/m2.32It is, thus, notsurprising that this problem generally appears onlyin patients being treated fo r m ore than 1 year,which requires a prolonged survival time.Compared to the other nitrosoureas, it can benoted that sem ustine nephrotoxicity totally differsfrom that of streptozotocin, the most nephrotoxiccompound of the nitrosourea group. The other ni-trosoureas. ie, chlorozotocin, lomustine, and car-mustine. seem to be much less nephrotoxic, withonly occasional cases re porte d.The specific mechanisms of semustine-relatednephrotoxicity remain unclear and there are noestablished recommendations for toxicity protec-t i on . P roch lo rpe raz ine , a commonly usedphenothiazine with antiemetic properties, has beenreported in animal studies to reduce the frequencyas well as the severity of semustine-related renallesions33: the clinical im pact of this observationhas not yet been established.

    STREPTOZOTOCINStreptozotocin is a nitrosourea that has beenused successfully in. the treatment of islet cellt u m 0 r s . ~ ~ - 3 ~enal toxicity is the major dose-limiting side"effect of this drug. Nephrotoxicity

    seems to be dcse-related and. to some extent, cu-mulative. occurring most frequently at doses in ex-cess of 1.5 g / m 2 / ~k .3 7~ 3aToxicity generally presents initially as mildpr~te inur ia . '~ut with continued u se, proximal re-nal tubular damage may dev elop.3 8 Fanconi syn-drome. renal tubular acidosis. hypokalemia, renalglycosuria. hypophosphatemia, and most signifi-cantly, life-threatening oliguria and anuria havebeen reported. Nephrogenic diabetes insipidusseems to be less frequent.*.41The best prevention for potentially harmfulnephrotoxicity should be afforded by an optimal

    dosage schedule and frequent examinations forp r o t e i n ~ r i a . ~ ~ . ~ ~he drug should be discontinuedwhen proteinuria or other signs of renal dysfu;: -tion develop; with resolving proteinuria, drug ..:. .-.ministration might be carefully continued.

    Mithramycin, a naturally occurring cytotoxicantibiotic, has shown some activity against go-nadal cancer, as well as g l i o b l a s t o m a s * ~ ~ ;ow-ever, its multiple toxic side effects have precludedits widespread use. Apart from causing importanthematologic and cloning abnormalities, gastroin-testinal, central nervous, and hepatic dysfunction,as well as hypoca lcemia, it also induces renal tox-icity, manifested by signs of renal failure and pro-teinuria.This renal toxic effect tends to be cumulat~:.and may be irreversible; drug-related deaths due ti:renal failure have been reported in 6 of 32 patientstreated with the drug.4s Histopathologic changesconsist primarily of renal tubular swelling, de-generation. necrosis, and atrophy with sparing ofthe glom eruli. M ithramycin still has some applica-tions in refractory cancer-related hypercalcemia,where lower doses are general ly eff ec t i~ e.* ~. ~ '

    Mitomycin C (MMC) is a naturally occurringalkylating agent, introduced for clinical use asearly as 1958. It has been demonstrated to be ac-tive against carcinomas of the gastrointest~n.:tract; breast. prostate, and a few other carcinomasas we11.49.s0Activation of mitomycin seems to require i nvivo metabolism to form short-lived metabolitesthat are highly reactive. Its serum half-life variesfrom nine to 17 minutes, and inactivation of thedrug occurs in various organs including the liverand the kidney. Renal excretion of mitomycin isnegligible.49Early reports on anima l studies had show n vari-ous nephrotoxic effects in dogs, monkeys, andmic e.s1-" Re ports on possible renal toxicity in hu-mans occurred only much later.s4 These studiesgenerally show ed m icroangiopathic changes in r h ~renal circulation with hem olytic anem ia. thrombo -cytopenia. proteinuria, hematuria, renal failure.systemic hypertension, pulmonary edema, andcongestive heart failure. This clinical syndromehas been variously described as microangiopathic

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    4/12

    NEPHROTOXICITY OF CMOSTATICS 371

    hemolytic anemia.55 MM C-related hemo lytic- tions without any effect on progressive renaluremic syndrom e.56 and MM C n ephrotox icity.57 f a i l ~ r e . ~ ~ . ~ ~ombinations of immunosuppressiveSeveral clinical manifestations show similarities and antiplatelet drugs, with or without plas-with cancer-related microangiopathy and coagu- mapheresis, has been shown to be only of marginal1 0 p a t h y , ~ ~ - ~ lnd different authors have suggested a benefit.63 Temporary progress ion of the diseasepossible relation between both problem^.^^.^^ activity has been attributed to blood transfusions,In one large series, 140 of 143 patients treated which should, the refo re, be administered withwith mitornycin had renal impairments7; among ~ a u t i o n . ~ ~ . ~ 'emodialysis may be necessary forthese, 6 % died of renal failure. Three patients de- life-threatening renal failure, but it will not effectveloped rapidly progressive renal disease with the underlying disease process.sharp elevations of BUN and'creatinine level. hy- For patients treated with MM C, renal and hema-penension, proteinuria, hematuria, signs of mi- tologic function should be monitored regularly af-croangiopathic hemolytic anemia. and death from ter initiation of chemotherapy. MMC should berenal failure. Several other patients in that series discontinued if early toxic manifestations, espe-presented with some degree of red-cell fragmenta- cially azoternia or hematuria, are detected. Totion, thrombocytopenia. proteinuria. and hyper- date, there are no data available to predict individ-tension, but the clinical course was generally less ual susceptibility to this complication. Furthertulminant and renal failure frequently developed studies are needed to clarify the causes, preven-without signs of hemolysis. As in other stud- tion. and treatment of the renal complication. andrelated to repeated doses of 10 to 20 mg lm2 , with a use.

    ., total dose-range between 40 and 115 mglm 2. Thetime interval between the onset of MMC therapy AZACYTIDINE: and renal failure was 4 to 16 months. with an 5-Azacytidine, a pyrimidine analogue, is a use-average of 10 to 11 months . ful agent in acute nonlymphocytic leukemia.70Renal toxicity of MMC is apparen tly not dose- This drug was show n to be associated with azote-related and the precise pathogenetic mechanism rnia and renal tubular d y ~ fu n c ti o n .~ ' bnormali-

    .: autopsy material have shown microangiopathic le- collecting ducts, have been described. Polyuria,sions with fibrous thrombi in the glomeruli and salt wasting, defective reabsorption of phosphate.' small arteries.65 glomerular nuclear degeneration aminoaciduria, and renal glycosu ria have also:. and thickened basement membrane.s4 intirnal hy- been d o ~ u m en te d .' ~

    CISPLATINIn 1969, platinum comp ounds were reported by

    . tients with MMC-associated hernolytic-uremic Rosenberg et al to exen potent antitumor activitysyndrom e63; these immune complexes showed in mice73; cisdiamm inedichloroplatinum I1 (cis-high platelet aggregation activity in vitro. T he platin) was the most active dru g amon g these com -constituent antibody of each complex faded to p o u n d ~ . ~ ~isplatin, therefore, entered toxicologicreact with MM C antigen preparations , whereas in evaluation in before introduction intovitro reactivity to endodermally derived neo- phase I clinical trial^.^^-^' Since then, cisplatin hasplasms could be detected. These data suggest that been studied in numerous phase 111111 trials (aloneMM C might not be the only etiologic factor of this or with other agents) , and it has proven t o be onesyndrome, but that some tumor-specific relation- of the most potent chemotherap eutic ag ents, beingship might exist as well. active in cancer of the ov ary ,8 4 t e ~ t i s , ~ . ~ ~lad-M itomycin -a ssoc ia ted nephrotoxic r eactions de r, 87 -89 ead and neck ,90 l ~ n g , ~ ' . ~ ~ndome-must be c~ns idered efrac tory to most trea tments t r i ~ m . ~ ~terine c ervix ,94 and many o thers.studied until now. In small studies. steroids, anti- Several excellent reviews deal with problem s suchf coagulants, and platelet inhibitors have been tried as mechanism of action, activity on diverse tu-( u n s u c c e ~ s f u l l y . ~ ~ ~ ~ ~ ~lasmapheresis has shown mors. as well as toxicity of isp plat in.^^.^^^I variable resolution of the hematologic manifesta- Since 1971, animal studies have shown a major

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    5/12

    RlES AND KLASTERSKY

    cisplatin-induced nephrot~xici ty'~hat proved tobe dose related.76 These and other reports withstudies performed on various animal modelsshowed a strong relationship between the adminis-tered dose and uremia, as well as dose-relatedstructural alterations detected on microscopic kid-ney examination. These alterations consisted pre-dominantly in tubular necrosis. As could be ex-pected from animal studies, early clinical reportsshowed similar toxicity in humans.78-m.82.a3n pa-tients with n&mal kidney function, rising BUNlevels were noted during the second week after abolus cisplatin administration in excess of 50 mg/m2 of body surface area (BSA); reversible nephro-toxicity was generally noted between 50 and 75mglm2ld, while doses 2 100 mglm21d were fre-quently followed by acute renal failure with pro-nounced tubular cell necrosis. Renal failure pro-gressing from reversible to irreversible stages wasdescribed with repeated treatment courses. Theseearly studies were performed without particularhydration or drug-induced diuresis programs.Several authors have reviewed the problem ofcisplatin-related nephrotoxi~ity~~~-~~~;he conclu-sions of their articles. as well as more recent infor-mation, will be analyzed in the following discus-sion. In fact, cisplatin nephrotoxicity remains ofparticular interest since renal damage is one of itsmost critical side effects and frequently constitutesa dose-limiting factor.Mechanism of Acrion and Implicationsfor Renal Toxici~

    Several studies suggest that cisplatin exerts itsactivity in a manner similar to aklylating agents bydrug interaction with \the nucleophilic sites ofpyrimidines in DNA.97.98.'06 hese infrastrandDNA cross-link will occur after physiologic ac-tivation of the dnlg, which will take place in a low

    u 1"UiCl , 103 .MIL1Fig 1. Chloridedependent aquation reactions ofcisplatin.

    chloride concentration environment: cisplatin pas-sage from a chloride-rich plasmatic milieu ( !03mEq/L) to a low-chloride invacytoplasmic mii;.. I(4 mEqIL) constitutes a passage from electr:.s.,lneutrality (with a stable bis-chloro molecule [I]) toaqueous activation (II, III, V), chloride ligands inthe cis position being replaced by water molecules(Fig 1). These aquated forms are highly reactivewith nucleophiles and can loose hydrogen ions toform cytotoxic hydroxyl radicals (IV, V); these lat-ter compounds can also form cytotoxic oxygen-bridged dimers and trimers. At equilibrium, theproportion between these diversely cytotoxic corn-plexes depends on chloride concentration and, to alesser extent, on pH and total platinum concentra-tion. 107.108

    Since platinum tissue concentrations are panl;larly high in the kidneys after drug admini~tratit~;~in dogs and hurnan~,~5.'~~nd data suggesting anactive tubular secretion of cisplatin or metabo-lites,'10-112t can be hypothesized that renal toxic-ity might be similar to its general cytotoxic effects.This implies that intratubular reduction of cisplatinconcentration by abundant hydration, as well asactivation of tubular chloride reuptake by forcedchlouresis, might favorably influence the equilib-rium between stable nontoxic cisplatin 0) andaquated forms (11, III, V), as well as cytotoxic hy-droxyl species (IV,VI), thereby reducing renal t u -bular damage without affecting general cytotoxic-ity.Cisplarin Tissue Disrriburion, Pharmacokinetics.and Renal Handling

    After IV administration. cisplatin undergoesrapid distribution to nearly all organs. Tissueplatinum concentration studies at various points af-ter cisplatin administration in dogs show a veryrapid accumulation of the metal in the kidney(three to four times the plasma concentration tenminutes after administration) with persistently ele-vated values fbr more than 1 week. op A very highpostmortem renal concentration of platinum hasalso been reported in humans two days after ad-ministration of a single dose of 2 mg/kg of ciiplatin.95 Platinum concentrates in various othertissues (gonadal tissue, lung, liver, fat)lo9;studiesin humans have shown that 90% of the drug isprotein-bound in plasma and that 27 % to 45% ofth e drug is excreted in the urine within five days ofadministration.11 3 Data on free platinum renalclearance in humans indicate that platinum clear-

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    6/12

    NEPHROTO XICITY OF CYTOSTATICS

    ance exceeds the GFR, suggesting active secretion[of the drug or a metab01ite.I~~ s might be ex--petted, cisplatin pharmacokinetics are altered ini atients with renal insufficiency, and normal ori near normal values of creatinine clearance are\generally considered necessary in order to avoidjcxcessive nephrotoxici@ This general rule seems, to be of questionable importance in cases of age-related decrease in renal function and especially incases of a unique functional kidney. In fact, bothconditions generally result in a lower 24-hourcreatinine clearance, but as has been shown in arecent clinical study, they are not associated withgreater renal toxicity if standard hydration is per-formed and if cisplatin dosage does not exceed 60mg/m2/d. I 4 This study even suggests a protectivemechanism in patients with unique kidneys (14 of43 patients studied) compared with those with twonormally functioning kidneys. In fact, the reducednumber of nephrons will produce a higher flow ofultrafiltrate per tubule with lower tubular cisplatinconcentration and probably less tubular "contact"between the toxin and tubular epithelium. This isillustrated by a higher than normal 24-hour diure-sis, wit!! lower cisplatin urinary concentrations foran identical cisplatin dose and hydration programin these patients, compared with those with two' ormally functioning kidneys. A similar mecha-nism is thought to contnbute to the relative lack ofexcessive age-associated cisplatin-induced nephro-toxicity.

    . Other studies (in animals and humans), based onrhythmic changes in kidney function, according tocircadian rhythms, have shown that optimal renaltolerance of cisplatin occurs very near to the timeof day associated with the normal circadian max-imum in urinary volume. This beneficial effect ispotentiated by concurrent hydration and is corre-lated by lower urinary cisplatin concentra-tions. 15- I l 7 These studies, along with those on hy-dration and forced diuresis suggest the primordialrole of reducing cisplatin urinary concentrationsby high urinary output; in fact. direct tubular ex-posure for a few hours to a concentration of 200pg/mL of cisplatin seems to be necessary for tubu-lar damage to occur.96Parhologic Kidney Changes

    As documented by animal and human studies.cisplatin induces several microscopic kidneychanges that are dose-related and, to a lesser de-gree. cumu!ative. 96,102, '03.118 These changes in-

    clude epithelial cell degeneration, proximal tubu-lar necrosis, dilatation and necrosis of distaltubules, interstitial edema. and lymphocytic in-filtration. Glomerular changes are generally notobserved and most alterations are considered to benonspecific. Tbbular damage is also suggested bysensitive assays for tubular enzymes, thought to beliberated by renal injury: alanine-aminopeptidase,N-acetyl-0-glucosaminidase (NAG) l 9 . I z 0 ;these studies support the concept that all areas ofthe tubule may be injured to some extent.

    Studies on urinary activity of NAG, a lysosomalenzyme released into the urine from the brush bor-der of proximal tubular cells, have shown a posi-tive correlation between cisplatin nephrotoxicityand NAG urinary activity. Potentiation of cisplatintissue uptake by NAG tissue activity has been re-

    as well as a circadian rhythm of urinaryNAG activity, suggesting that rhythmic changes inNAG activity may cause variations in cisplatinnephrotoxicity along with circadian rhythms. PZ -microglobulin, possibly another mediator of tubu-lar injury, was shown to be significantly elevatedin the urine 12 hours after treatment with cis-platinI2l: since no patient in that report developedlaboratory evidence of renal toxicity, this in-creased /32-microglobulinuriahas been interpretedas reflecting subclinical renal damage.Hypomagnesemia and renal magnesium wastingare the most prominent electrolytic disorders sec-ondary' to cisplatin administration. 1 2 2 - 1 2 4 Hypo-magnesemia d e v e l o e 2 % of 44 patie-h e eriesLz2 ith a treatment schedule of 70 mg/m2 cisplatin (on day l ) , repeated every 21 days.Among patients with hypomagnesemia. the nadirMg concentration (median) was 0.92 mEq/L.usually occumng more than 2 weeks after drugadministration and generally after one or twocourses of cisplatin had been given. Similar hypo-magnesemia was also observed in other stud-ies1Z3.125nd is frequently complicated by hypo-calcemia that is probably secondary to diminishedPTH release and/or end-organ resistance toparathyroid hormone induced by hypomagnese-mia.126z 7 Clinical manifestations of hypomagne-semia. including abnormalities in neuromuscular.central nervous system. and cardiac function thatmay appear for serum levels of less than 1 mEq/L.should be treated by parented replacement ofmagilesium sulfate. Associated hypocalcemia isresponsive to .magnesium repletion and is unre-sponsive to calcium replacement alone. The more

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    7/12

    RlES AND KLASTERSKYrarely seen hypokalemia should be treated by re-placement of both potassium and magnesium.Prevention of'Cisplatin-Induced Nephrotoxiciry

    Preexistent renal insufficiency should always beexcluded before starting cisplatin therapy; dehy-dration, arterial hypeitension and hyperuricemiashouid be corrected; concomitant administrationof nephrotoxic drugs, especially aminoglycosideantibiotics, should be avoided.Iz8Numerous clinical trials proposing various hy-d ra tio n p ro gr am s h av e been d e ~ c r i b e d . ~ ~ . ~ ~ . l ~ 9llof these schedules are valuable, provided that hy-dration status is closely mon itored. Most hydrationprograms are performed with normal saline solu-tion or glucose 5% in saline and are given for vari-ous durations, before, during, and after cisplatinadministration. Hydration alone can be consideredsufficiently effective if cisplatin is administered as20mglm2ld by one- to two-hour infusion for fiveconsecutive day s.

    Forceddiuresis programs, combining hydrationwith furosemide or m annitol administration, haveproved clinically beneficial in numerous stud-ies,84.130.132nd superiority of hydration-diureticprograms compared to hydration alone has beensuggested.'33 Most of the more recent programspermit administration of cisplatin at doses of 100ro 120mg /m2 as short daily infusions or adminis-tration of 20 o 40 mglm21d for five consecutived a y s . I n t e n s iv e p a r e n t e r a l h y d r a t i o n w i thmannitol-induced diuresis even permits preventionof dose-limiting nephrotoxicity. In these patients.myelotoxicity might become a major problem134;combined with cisplatin administration in hyper-tonic saline, doses as high as 200mgl m2 have beenadministered, with thrombocytopenia being themos t promineht toxic effect."' In a recent study,three "hig hdo sf" regimens, with doses between180 and 220 mg lm2 /d, were compared: cisplatinwas dissolved in 5% saline and was administeredalong with a well-defined prehydration-mannitol-induced diuresis and posthydration program. Inthat study it was concluded that cisplatin, given ata dose of 200mg /m2 in five daily fractions, com-bined with an appropriate hydration and diuresisprogram. was devoid of significant nephrotoxicityand was probably much less ototoxic than single-bo lus h igh-dose admin i~ t ra t i0n . l~~igh chloridediure sis may improve the therapeutic index of cis-platin by reduced nephrotoxicity without loss ofantitumor activity. Animal studies have shown in-

    creased nephrotoxicity in chloridedeprived ratsand significantly reduced renal damage with phar-macologically induced chloriuresis; this favorabi:effect could be explained by the reduction nihighly toxic aqueous-cisplat in complexes inchloride-rich urine. lo 'Continuous infusion of cisplatin results in re-duced gastrointestinal and renalwhen compared with earlier studies with equaldaily doses. C ontinuous infusion should, however,always be asso ciated with adequate hydration pro-grams, the influence on renal toxicity of vigorousurinary output being more important than theduration of infusion.Pharmacologic Interaction WithCisp la t in Nephro tox ic i~

    Unlike mannitol and furosemide, which exerttheir activity by inducing a potent diuresis, variousother compounds have been proposed on the basisof a potential for detoxification. Among these.probenecid has been studied in animals (F344 atmodel) and w as shown to decrease the peak valuesof BUN and creatinine if administered one hourprior to cisplatin; this protection has been ascribedto probenec id inhibition of activ e secretion of cis-platin by the p-aminohippu rate system.14'WR-2721 S-2-(3-aminopropy1amino)-ethylphosphorothioic acid] , a radioprotect iveagent, has been shown to exert protective activityon renal function in the same F344 rat model.IJ2Diethyldithiocarbamate (DDTC). a potent SH-group chelator administered to F344 rats after aknown nep hrotoxic dose of cisplatin. may preventrenal pathologic a s well as functional changes.14'This protective activity is ascribed t o comp etitionwith cisplatin for protein-bound sulfhydril groupswithin proximal tubular cells. Protein binding ofcisplatin in tubular cells, with resultant inhibitionof transport enzymes, is one postulated mecha-nism of cisplatin nephrotoxicity in rats and,perhaps, in humans. The closely related disulfiram(An tabus e: Ayerst Laboratories. N ew York). beingmetabolized to diethylthiocarbarnate. might exen asimilar protective effect.

    Sodium thiosulfate has also been studied for itsnephrotoxicity preventive effects. L44 T h i s s u lh r -containing compound is thought to reacr CO -valently with cisplatin, resulting in com plexe s thata re n eith er toxic n or t ~ m o r i c i d a l . ~ ~ ~part frommarkedly reducing general cisplatin toxicity by itssystemic activity, sodium thiosulfate might exert

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    8/12

    PERSPECTIVES AND CONCLUSIONSA better knowledge of various factors contribut-

    ing to cisplatin nephrotoxicity, as well as optimaladministration programs with hydration and dmg-

    further reduction in nep hrotoxicity through partic-ularly high intratubular thiosulfate to cisplatin c on-centration ratios. Intravenous sodium thiosulfateseems to be a most intiresting agent when ad-ministered in combination with intraperitoneal cis-platin. eg, for ovarian cancer. In this indication.parenteral thiosulfate adniinistration allowed e sca-lation of intraperitoneal cisplatin up to a dose of270 mg /m2 without significant nephrotoxicity, thuspermitting an increased activity.i4s Other sulfurcompounds, eg, thiourea, L-methionine. and peni-cillarnine, might also inhibit the biologic activity

    induced diuresis, have considerably changed the

    of cisplatin and might, therefore, interfere with itstherapeutic a s well as toxic effects.1 4C 148The carbonic anhydrase inhibitor acetazolarnidehas also been considered potentially useful for p re-vention of cisplatin nephrotoxicity. 49.1s0 APa*from its activity as a diuretic, this organic acidcould act by com petition with cisplatin for tubularreabsorption, thus promoting cisplatin excretion:however, the presence of two sulfur atoms in thismolecule could also permit chemical interactionand detoxification. In an animal study (male F 344rat model), pretreatment with acetazolarnide de-creased cisplatin nephrotoxicity and diminishedrenal platinum content, as well as urinary platinume ~ c r e t i 0 n . l ~ ~omparison of acetazolamide andmannitol i n the same animal model (perform ed bythe same authors) shows acetazolamide to be mo reeffective than mannitol in preventing cisplatin-induced nephrotoxicity in male F344 rats. l S 0 Theenzyme copper-zinc superoxide dismutase (orgo-tein) has also been associated with reduction ofnephrotoxicity in rats, thus suggesting that su-peroxide radicals may participate i n the nephro-to xic effe cts of c i ~ p l a t i n . ~ ~ ~Selenium. known for its ability to combine tometals such as cadmium and mercury, as well asfor its resem blance to sulfur by its chem ical prop -erties, shows theoretical promise as a cisplatin-precipitating agent. As shown in studies on mice.selenium is able to decrease cisplatin neph rotoxic-ity without influencing in vivo and in vitro tumorgrowth. This could suggest either detoxificationby ligation to sites not involved in cisplatin antitu-mor activity or, perhaps, prevention of tubularreabsorption of the mo1esu:ar complex. 1 5 2

    spectrum of cisplatin's potential for renal toxicity.Introduction of some of the new detoxificationagents will probably prove useful as well. Dosingplasma cisplatin levels at various times after druginfusion could be helpful to predict nephrotoxicityearly in the course of cisplatin infusion . I s3 Finally,several second-generation platinum com plexes arebeing evaluated in preclinical and clinical studiesfor toxicity and antitumor activity. Among these,i p r o p l a t i ~ u m ( C H I P ) a n d c a r b op l a ti n u rn(C BD CA ) show l ittle or no n e p h r o t o x i ~ i t y . ~ ~ ~ - ~ ~ ~These and other platinum com poun ds are possiblycandidates as future alternatives to cisplatin inseveral conditions. Until then. however, cisplatinremains one of the most potent antineoplasticagents ever developed. Further work should beperformed to reduce its potential for renal toxicity.

    REFERENCESI. Garnick MB. Mayer RJ: Management of acute renal

    failure associated with neoplastic disease. in Yarbm J. Born-stein R (eds): Oncologic Emergencies. Management of AcuteRenal Failure Associated With Neoplastic Disease. Orlando.FL. Grune & Stranon, 1981, pp 247-2712. Weiss RB. Poster DS: The renal toxicity of cancer che-motherapeutic agents. Cancer Treat Rev 9:37-56. 1982

    3. Abelson HT.Garnick MB: Renal failure induced by can-cer chemotherapy, in Rieselbach RE. Garnick M B (eds): Can-cer and the Kidney. Philadelphia. Lea & Febiger. 1982. pp769-8 13

    4. Bleyer WA: The clinical pharmacology of methotrexate:New applications of an old drug. Cancer 4 1 :36-5 1 . 19785. Frei E UI. Jaffe N. Tanenall MHN. et al: New ap-proaches to cancer chemotherapy with methotrexate. N Engl JMed 292:846-85 1. 19756 . Pran CB. Roberts D. Shanks E. et al: Response. toxicity.and pharrnacokinetics of highdose methotrexate (NSC-740)

    with citrovorum factor (NSC-3590) rescue for children withosteosarcorna and other malignant tumors. Cancer ChemotherRep 6: 13- 18. 1975 (suppl)

    7. Stoller RG. Jacobs SA. Drake JC. et al: Pharmaco-kinetics of highdose methotrexate (NSC-740). Cancer Che-mother Rep 6: 19-24. 1975 (suppl)8. Von Hoff DD. Penta JS. Helman U. t al: Incidence ofdrug-related deaths secondary to highdose of methotrexate andcitrovorum factor administration. Cancer Treat Rep 61:745-748. 1977

    9. Pitman SW. Parker LM. Tattersall MHN. et al: Clinicaltrial of highdose methotrexate (NSC-740) with citrovorumfactor (NSC-35%)-Toxicologic and therapeutic observations.Cancer Chemother Rep 6:43-49. 1975 (suppl)

    10. Bratlid D. Moe PJ: Pharmacokinetics of highdosernethotrexate treatment in children. Eur J Clin Pharmacol14: 143-147. 1978I I. Pitman SW. Fr e~ 111: Weekly rnethurexatetalciumleucovorin rescue: Effect of alkalinization on nephrotoxicity;

    pharn~acokineti:~n the CNS: and u x in CtJS non-Hodgkin'slymphoma. Cancer Treat Rep 61 :695-701. 1977

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    9/12

    12. Frei E Ill. Blum RH. Pitman SW. et al : H i g h d o s emethotrexate with leucovorin rescue. Rationale and spectrumof antitu mo r activity. Am J Med 68:3 70-3 76. 198013. Rom olo JL. Goldb erg NH. H ande KR. t al: Effect ofhydration on plasma methouexate levels. Cancer Treat Rep

    61: 1393-13%. 197714. Pinedo HM. Chabner BA: Role of drug concentration,durationaf exposure, and endogenous metabolites in determin-

    ing methotrerate cytotoxicity. Cancer Treat Rep 61:709-775.1977

    15. Link DA. Fosburg MT. Ingelfinger JR. et al: Renal tox-icity of highdose-m ethouexate. Pediatr Res 10:455. 1976 (ab-s a 92 3)

    16. Howell SB, Carmody J: Changes in glomerular filtrationrate associated with highdose methotrexate therapy in adults.Can cer Treat Rep 6 1: 1389- 1381. 197717. Abelson HT. Fosburg MT. Bea rdsley GP. et al:Methotrexate-ind uced renal im pairment: C linical studies andrescue from systemic toxicity with highdose leucovorin andthymidine. J Clin Oncol 1:208-216. 1983

    18. Tanersall MHN. Parker LM. Pitman SW. et al: Clinicalpharmacology of highdose methotrexatc. Cancer ChemotherRep 6:25-29. 197519. lsacoff WH. Townsend CM. Eibcr FR, et al: Highdosemethotrexate therapy of solid tumors: Observations relating toclinical toxicity. Med Pediatr Oncol 21319-325. 1976

    20. Ahmad S. Shen F. Bleyer WA: M ethotrexatc-induced re-nal failure and ineffectiveness of peritoneal dialysis. Arch In-tern Mcd 138:1146-1147. 1978

    2 1. Hande KR. Balow JE. Drake JC. et al: M ethotrexate andhemodialysis. Ann Intern Med 87 :49 54% . 197722. Gibson TI? Reich SD. Krum lovsky FA. et al: Hemo per-fusion for methotrexate removal. Clin Pharmacol Ther23:351-355. 197823. Djerassi I. Ciesielka W, Kim JS: Removal of methotrex-

    ate by filtration-adsorpt~on sing charcoal filters or by hemo:dialysi s. C ancer Treat Rep 61 :75 1-752. 197724. Oliverio VT:Toxicology and pharm acology of the nitro-soure as. Cancer Chemother Rep 4: 13-20. 197325. Denine EP, Hamson SD Jr. Peckham JC: Qualitativeand quan titative toxicity of sublethal doses of methy l-CCNU in

    BDF , mice. Cancer Treat Rep 61:4 09-4 17. 197726. Schacht RG. Baldwln DS: Chronic interstitial nephritisand renal failure due to nitrosourea (N U) therapy. Kidney Int

    14:661. 1978 '27. Harmon WE. Cohen HJ. Schneegerger EE. et al:Ch ron ic renal fai1u;e in children treated with me thyl-C CN U. N

    Engl J Med 300: 1200-1203. 197928. Schacht RG. Feiner HD. Gallo GR. et al: Nephrotoxic-ity of nitros wrea s. Cancer 48: 1328- 1334, 198 129. Nichols WC. ~MoertelCG: Nephrotoxicity of methyl

    CCN U. N Engl J Med 301:1181. 1979 (lener )30. Kundaria 8. Armentrwt S. hlargileth D. et al: Nephro-

    toxicity of semustine. Proc Am Soc Clin Oncol 223352. 1981(abstr C-77)

    31. Micetich KC. Jensen-Akula M. Mandard JC. et al:Nephrotoxicity and semustine (methyl-CC NU) in patients withmalignant melanoma receiving adjuvant chemotherapy. Am JMed 7 1 967-972. 1981

    32. Weiss RB. Posada JG. Kram er RA. et al: Neph rotox icityof semustine. Cancer Treat Rep 67: 1 105-1 1 12. 1983

    R lES AND KLASTERSKY33. Harrison SD Jr. Cox JL, Giles RC Jr: Effbcu of pro-

    chlorpcrazine on experimental nephrotoxicity. Cancer Che-mother Pharma col 10:62-67. 198234. Taylor SG Ill. Schwanz TB. annini JJ, et al: Streptoz,tw in therapy for metastatic insulinoma. Arch Intern .\I

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    10/12

    NEPHROTOXICITY O F CYTOSTATICS- 55. Gulati SC, Sordillo P Kem p~n . et al: Microangtopath~c

    hemolytic anerma observed after treatment of epldermoid car-'? cinoma wirh Mitomycin C and 5 -f luo rouraci l . Cancer

    45:2252-2257. 1980 .56. Pavy MD. Wiley EL. Abeloff MD: Hemolytic-uremicsyndrome associated with mitomycin therapy. Cancer Treat

    R ep 6 6 : 4 5 7 4 1 . 1 98 2 '.,

    57. Hanna W. Krauss S. Regester RF. et al: Renal diseaseg after mitomycin-C therapy. Cancer 48:258 3-2588 . 1981X 58. Brain MC, Azzopardi JG. Baker LRI. el al: Microan-

    giopathic hemolytic anemia and mucin-forming adenocar-#: cin om a Br J Haematol 18: 183-192. 197059. Lohr mann H. Adam W. Hcymer B. et al: Micro an-giopathic hemolytic anemia in metastatic carcinoma. Ann In-tern Med 793 68-3 75, 1973

    60. Sack GH. Levin J. Bell WR: Trousseau's s ynd rom e andother manifestations of chronic dissermnated coagulopathy inpatients with neoplasms: Clinical, pathophysiologic, and thera-peutic feawres. M edicine 56: 1-37. 1977

    61. Antman KH. Skarln AT. Mayer RI. et al: Microan-giopathic hemolytic anemia and cancer. A review. Medicine58:377-384. 1979

    62. Kressel BR. Ryan KP. Duong AT. et al: Microan-giopathic hemolytic anemia. thrombocytopenia and renalfailure in patients treated for adenocarcinoma. Cancer48:1738-1745. 1981

    63. Cantrell JE, Phillips TM , Schein PS: Carcinoma-associated hemolytic-uremic syndrome: A complicatton of mi-l ' tomycin C chemotherapy. J Clin Oncol 3~7 23-7 34. 1985J 64. Krauss AS. Sona da T. Soloman A: Treatment of ad-vanced gastrointestinal cancer with 5-fluorouracil and mitomy-cin C. Cancer 43: 1598-1603, 1979

    55 . Ham ner RW. Verani R. Weinman EJ: Mitomycin-associated renal failure. Arch Intern Med 143:803-807. 198366. Jones BG. Newman CD. Fielding JW. et al: Intravascu-lar haemolvsis and renal impairment after blood transfusion inrwo patients on long-term 5-fluorouracil and mitomycin C.

    1 Lancet 1: 1275-1 276. 198067. Rumpf KW. Rieger J. Lankisch PG. t al: Mitomycin-i induced haem olvsis and renal failure. Lancet 2: 1037- 1038.1980I 68. Ratanatharathorn V Baker LH. Cadoapaphorchai P. rtal: Clinical and pathological swdy of mitomycin C nephrotox-icity. in Caner SK. Crooke ST (eds): Mitomycin C. Current( S I ~ N Snd New Developments. Orlando. FL. Academic 1979.I pp 2 19-22969. Lempen KD: Haemolysis and renal impairment syn-

    drome in patients on 5-fluorouracil and mitomycin C. Lancet2:369-370. 1980I 70. Von Hoff DD. Slavik M. Mugg ia FM: 5-Azacy tidine: A1 new anticancer drug with effectiveness in acute myelogenousI leukemia. Ann Intern Med 85:237. 197671. Greenberg MS: Reversible renal dysfunction due to 5-I azacytidinc. Cancer Treat Rep 63:806. 197972. Peterscn BA. Collins AJ. Vogelzang NJ. et al: 5 -Azacytidine and re r~a l ubular dysfu nction. Blood 57: 182-:85.198 17 3 . ~ o s e n b e ; ~. Van Camp L. Trosko JE. ct al: Platinum

    compounds. Saw re 22238 5-386. 196974 . Ro se nk rg B. Van Camp L: The successful regrcsson of

    large solid sarcoma 180 tumors by platinum com pound s. Can-cer Res 30: 1799- 1802. 1970

    75. Kociba RJ. Sleight SD: Acute toxicologic and patho-logic effects of cisdiamrninedichloroplatinum (NSC-119875)in the male rat. Cancer Ch emother R ep 55: 1-9. 197176. Leonard BJ. Eccleston E. Jones D. et al: Antileukaemicand nephrotoxic propenies of platinum compounds. Nature234:43-45. 1971

    77. Schaeppi U. Heyman 1A. Flcischman RW. et al: Cis-dichlorodiammineplatinum(IJ) (NSC-I 19 875): Preclinical tox-icologic evaluation of intravenous injection in dogs. monkeysand mtce. Toxicol Appl P h a m c o l 25:230. 1973

    78. Rossof AH. Slayton RE. Perlia CP: Prelimin ary clinicalexperience with cis-diamminedichloroplatinum (11) (NSC-1 19875. CACP). Ca ncer 30: 1451-1456. 1972

    79. Lippman AJ. Helson C. Helson L. et al: Clinical trialsof cis-diamrninedichloroplatinum (NSC -I 19875). Cancer Che-mother Rep 57:191. 1973

    8 0 . H ig b y D J . W al l ace H J J r. H o l l an d JF : C i s -diamm~nedichloroplatinum (NSC-1 19875): A phase I study.Cancer Chemother Rep 57:459-463. 1973

    81. Talley RW. O'Bryan RM. G utterman JU . et al: Clinicalevaluation of toxic effects of cisdiamminedichloroplatinum(NSC-119875): Phase I clinical study. Can cer Chemother Rep57:465-47 I, 1973

    82 . De Conti RC. Toftness BR. Lange RC. et al: Clinicaland pharmaco log ical s tud ies wi th c is-d iammined ich lo ro -platinum 111). Can cer Res 33:1310. 197 3

    83. Higby DJ. Wallace HJ Jr. Alb en DJ. et al: Diammino-dichloroplatinum: A Phase I study show ing respon ses In testic-ular and other tumors. Cancer 33:1219-1225. 1974

    84. Young RC, Von Hoff DD. Go rmley P. et al: Cis-diamminedichloroplatincm (11) for the treatment of advancedovarian cancer. Cancer Treat Rep 63: 1539- 1544. 19798 5 . E in h o rn L H . D o n o h u e J : C i s -d i am m in ed ich lo ro -platinum. vinblastine and bleomycin combination chemother-apy in disseminated testicular cancer. Ann Intern Med 87 :293-298. 1977

    86. Garnick MB. Canellos GP. Richie JP. et al: Sequentialcombination chemotherapy and surgery for disseminated testic-ular cancer: Cisdiamminedichloroplatinum. vinblastine andbleomycin remission-induction follow ed by cyclophosphamideand adriamycin. Ca ncer Treat Rep 63 : 1681 1686. 1979

    87 . Yagoda A: Phase 11 t r ia ls w i th c is-d iam mined i-chloroplatinum (11) in the treatment of urothelial cancer. Can-cer Treat Rep 63: 1565- 1572. 1979

    88. Williams SD. Donohue JP. Einhorn LH: Advanced blad-der cancer: Therapy with cisdiamminedichloroplatinum (11).ddriamycin and 5-fluorouracil. Cancer Treat Rep 63:1573-1576. 1979

    89. Samuels ML. et al: CISCA combination chemotherapyfor metastatic carcinoma of the bladder. in Johnson DE. Sam-uels ML (eds) : Cancer of the Genito urinary Tract. New York.Raven. 1979. p 10190. Hong WK. Bhutani R. Shapshay SH. el al: Induction

    chemotherapy of advanced previously untreated squamous cellhead and neck cancer with cisplatin and ble om )c~ n. in Pre-stayko AW. Cr wk e ST. Carter SK (eds ): Cisplattn-CurrentSratus and New Developments. Orland o. FL . .4cademic. 198 0.p 431

    91 . Eagsn RT: Phase I1 study of cyclophosph amide.

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    11/12

    RlES A N D KLASTERSKadriamycin, and cisdiamrninedichloroplatinum (11) by infusionin patients with ademcarcinoma and large cell carcinoma of thelung. Cancer Treat Rep 63: 1589. 197992. De Jaeger R, bng ev al E. Klastersky J: Highdose cis-ptatin with fluid and rnannitol-induced diunsis in advancedlung cancer: A phase U clinical trial of the EORTC Lung Can-cer Working Party e lg ium) . Can ar Treat Rep 64: 1341-1346, 198093. Baker LH: Cisplatin in treatment of cervical and endo-metrial cancer patients. in Rcstayko AW Crooke ST, CarterSK (eds): Cisplatin-Current Status and New Developments.Orlando. FL. Academic. 1980. p 403

    94. Th ie pn IT. Shingleton H. Homesley H. et al: Cisplatinin the treatment of advanced or recurrent cervix and uterinecancer. in Prestayko AW Crooke ST. Caner SK (eds):Cisplatin-Current Status and New Developments. Orlando.FL. Academic. 1980. p 41 1

    95. Hill JM, LoebE, MacLcllan A, et al: Clinical studies ofplatinum corodination co m p n d s in the treatment of variousmalignant diseases. Cancer Chemother Rep 59:647-659. 197596. Rozencweig M. Von Hoff DD. Slavik M, et al: Cis-diamminedichloroplatinum (In. a new anticancer drug. Ann In-tern Med 86:803-812, 197797. Einhorn LH, Williams SD: The role of cis-platinum insolid tumor therapy. N Engl J Med 300:289-291. 197998. Zwelling LA , Kohn KW: Mechanism of action of cis-diamminedichloroplatinum (11). Cancer Treat Rep 63:1439-1444. 1979

    99. Guarino AM, Miller DS. Arnold ST, et al: Platinate tox-icity: Past. present, and prospects. Cancer Treat Rep 63: 1475-1483. 1979100. Prestayko AW CrookeST. Cancr SK (eds): Cisplatin-Current Status and New Developments. Orlando. FL. Aca-demic. 1980101. Hardaker WT Jr. Stone RA. McCoy R: Platinumnephrotoxicity. Cancer 34: 1030-1032. 1974102. Gonzalu-Vitale JC. Hayes DM, Cvitkovic E. et al:The renal pathology in clinical trials for cis-platinum (II)diam-rninedichloride. Cancer 39: 1362- 1371. 1977103. Madias NE, Hanington JT: Platinum nephrotoxicity.Am J Med 65:307-314. 1978104. Stark JJ. Howell SB: Nephrotoxicity of cis-platinum

    (11) dichlorodiammine, Clin Pharmacol Ther 23:461-466.1978105. Comis RL: Cisplatin nephrotoxicity: The effect ofdose. schedule. and hydration scheme. in Prestayko AWCrooke ST. Carter SK (cds): Cisplatin-Current Status andNew Developments. Orlando. FL. Academic. 1980. p 485106. Rosenberg B: Cisplatin: Its history and possible mecha-nisms of action. in Prestayko AW Crooke ST. Gn e r SK (eds) :Cisplatin-Current Status and New Developmenrs. Orlando.FL. Academic. 1980. p 9107. Lee KW Martin DS: Ci~di~hlorodiamrnine~latinum(ID. Aquation equilibria and isotop~c xchange of chloride l i-gands with free chloride and tetrachloroplatinate (11). lnorpChim Acta 17:105-110. 1976108. Earhan RM, Martin PA. 'htsch KD. et al: Improve-ment in the therapeutic index of cisplatin (NSC 119875) bypharmacologically induced chloriuresis in the rat. Cancer Res43: 1 187-1 194. 1983109. Le Roy AF. Lutz RJ. Dedrick RL. et al: Pharmaco-

    kinetic study of cis4chlorodiammineplatinum (XI) (DDP)the beagle dog: Thermodynamic and kinetic behavior of DTin a biologic milieu. Cancer Treat Rep 6359-71, 1979110. Jacobs C, Kalrnan SM. Treaon M, et al: Rend h:dling of cisdichlodiammineplatinum0. ancer Trcat Rt64:1223-1226. 1980I I I . PattonIT, immelstcinKJ. Belt R. et al: Plasma level.and urinary excretion of filterable platinum species followin!bolus injection and i.v. infusion of cis-dichloro.diamrnineplatinum (I0 n man. Cancer Treat Rep 62~1359-1362, 1978112. Gonnley PE, Bull JM, LeRoy AF, et al: Kinetics of cis-dichlorodiammineplatinum.Clin Phannscol Thcr 25:351-357.1979113. Frick GA. Ballentine R.Driever CW. t al: Renal ex-cmion kinetics of highdose cirdichlorodiammincplatinurn(II) administered with hydration and manniwl diurcsis. CancerM t ep 63: 13-16, 1979114. Hrushesky WJM. Shimp W Kennedy BJ:Lack of arcdependent cisplatin nephrotoxicity. Am J Mcd 76:579-5.-1984115. Hrushesky WJM. Levi FA. Halkrg F, et al: Cisplatinlethal toxicity is circadian stagedepcndent. Cancer Res42:945-949, 1982

    116. Levi FA, Hrushesky WWJ, Blomquut CH: Reductionof cisplatin nephrotoxicity by optimal circadian drug timing.Cancer Res 42:950-955. 1982117. Levi FA, Hrushesky WJM, Borch W, t al: Cisplatinurinary pharmacokinetics and nephmtolicity: A common cir-cadian mechanism. Cancer Treat Rep 66: 1933- 1938. 1982118. Agganval SK. Whitehouse MW, Rvnachandran C: U1-trastructural effects of cisplatin. in Restay ko AW Crooke ST.Caner SK (4 s): Cisplatin. Orlando. FL. Academic, 1980. p79119. Jones BR, Ravi BB. Mladek J. et al: Comparison ofmethods of evaluating nephrotoxicity of cis-platinum. C1.'-1Pharmacol Ther 27557-562. 1980120. Diener U. Knoll E. Langer B,et 81: Urinary excretionof N-acetyl-8-glucosaminidasc nd alanine arninopcptidase inpatienu receiving amikacin or cis-platinum. Clin Chim Acta112:149-157, 1981121. Cohen AL. Hargerg J, Citrin DL: Measurement of

    urinary 6, microglobulin in the detection of cisplatin nephro-toxicity. Cancer Treat Rep 65:1083-1085. 1981122. Schilsky R, Anderson T: Hypornagnexmia and ma 1magnesium wasting in patienu receiving cisplatin. AM InternMed 92929-93 1, 1979123. Anderson T, Javadpour N, Schilsky R, et al: Chemo-therapy for tes t~cular ancer: Current status of the NationalCancer Institute combined modality trial. Cancer Treat Rep63~1687-1695. 1979124. Blachley JD. Hill JB: Renal and electrolyte disturb-ances associated with cisplatin. Ann Intern Med 95:628-6.':.1981125. Hill JB, Blachley JD. Trotter M: Hypomagnesemla.hypcalcemia, and hypokalemia with cis-platinum treatmentClin Res 26:780A, 1978 (abnr)126. Suh SM, Tashjian AH Jr. Mauuo N, et al: pathogenesisof hypocalcernia in primary hypomagnescmia: Normal end-organ responsiveness to parathyroid hormone impairedparathyroid gland function. J Clin Invest 52:153-160. 1973

  • 8/2/2019 09 Nephrotoxicity Induced by Cancer Chemotherapy With Special Emphasis on Cisplatin Toxicity

    12/12

    127. Connor TB.Toskes P, Mahaffey J. et al: Parathyroidfuncrion during chronic magnesium deficiency. Johns H opluns\fed J 131:100-117, 1972

    1 2 8 . Ha a s A . ~n d e r s o nL. Lad T: The influence ofaminoglycosides on the nephrotoxicity of cis-dichloro-diammineplatinum in cancer patients. J Infect Dis 147:3634,1983129. Salem P.Hall SW. Benjamin RS. et al: Clinica l phase I-II study of cisdichlorodiammine platinum (ID given by con-tinuous IV in hi on . Cancer Treat Rep 62:1553-1555. 1978130. Chary KK, Higby DJ. Henderson ES. et al: Phase 1study of high dose cisdichlorodiammineplatinum (iI) ithforced diuresis. Cancer T m t Rep 6 1:367-370. 1977

    131. Merrin C. Beckley S, Takita H: MultimoQl treatmentof advanced testicular tu M r with radical reductive surgery andmultisequential chemotherapy with cis-platinum. bleomycin.vinblastine. vincristine and actinomycin D. J Urol 120:73-76.1978

    132. Hayes DM. Cvitkovic E.Golbey RB. t al: High dose,-1s-platinumdiammine dichloride. Amelioration of renal toxic-:ty by mannitol diuresis. Cancer 39: 1372-1 381. 1977133. Al-Sarraf M. Fletcher W Oishi N. et al: Cisplatin hy-dration with and without mannitol diuresis in refractory dis-seminated malignant melanoma: A Swthwcs t Oncology G roupstudy. Cancer Treat Rep 66:3 1-35, 1982

    134. Chary KK. Higby DJ. Henderson ES. et al: Phase Istudy of high dose cisdichlorodiammineplatinum (11) withforced diuresis. Cancer Treat Rep 61:367-370. 1977135. Ozols RF. Corden BJ. Jacob J. et al: High dose cis-platin in hypeno nic saline. Ann Intern Med 100: 19-24, 1984136. Blumemich MS. Woodcock TM. Jones M. et al:Highdose cisplatin in patients with advanced malignancies.C.mc er 5 5: 11 18-1 122. 1985137. Jacobs C. Benino JR, Goffinet DR. et al: 24-hour infu-sion of cis-platinum in head and neck cancers. Cancer42:2135-2140. 1978

    138. Kuhn JA. Argy Wi? Barnard W et al: Lessening of cis-dichlorodiammineplatinum nephrotox~ciryby protracted infu-sion. Clin Res 28:837A, 1980 (abstr)1 3 9 . Lo k i c h J J : Ph a s e I s t u d y o f c i s - d i c h l o r o -diammineplatinum (11) administered as a conswt 5-day infu-sion. Cancer Treat Rep 64:905-908. 1980

    140. Carlson RW, Sikic BI: Conttnuous infusion or bolusinjection in cancer chemotherapy. AM Intern Med 99:823-833. 1983141. Ross DA. Gale GR: Reduction of the renal toxicity of

    cis-dichlorodiammineplatinum (11) by probenecid. CancerTreat Rep 63:781-787. 1979142. Yuhas JM. C ulo F: Selective inhibition of the nephro -toxicity of cisdichlorodiammineplattnum (LI) by WR-27-21without altering its antitumor propenies. Cancer Treat Rep64:57-64. 1980

    143. Borch RF. Pleasants ME: Inhibition of cis-platinum

    nephrotoxicity by diethyldithiocarbamatc rescue in a rat model.R oc Natl Acad Sci 76 56 1 1-6614. 1979144. Howell SB. Taetle R: Effect of sodium thiosulfate oncisdichlorodiammineplatinum (11) oxicity and antitumor ac-tivity in L1210 leukemia. Cancer Treat Rep 64:611-616. 1980145. Howell SB. Pfeif le CL. Wung WE, et al : Intrapri teneal cisplatin with systemic thiosulfate protection. AM internMed 97:845-851. 1982146. Filipski J. Kohn KW. Prather R. et al: Thiourea m-

    verses cross-links and restores biological activity in DNAtreated with dichlorodiammineplatinum (LI). Science 204: 181-183, 1979147. Newman AD. Ridgway H. Spoor RI. et al: Inhibitionof biological activity of cisplatin by thiourea and L-methionire.J Clin Hematol Oncol 9:208-209. 1979148. Higby DJ. Wallace HI Jr. &kcsi JG : Reduction of cis-dichlorodiammineplatinum U toxicity by pe nicillamine com-pounds in animal models and humans. Proc Am Assa: CancerRes 16:131. 1975149. Osrnan NM. Copley Mi? Litterst CL: Amelioration ofcisplatin-induced nephrotoxicity by the diuretic acerazolamidein F344 rats. Cancer Treat Rep 68:999- 1004. 1984150. Osman NM. Copley MI? Lincnt CL: Effms of thediumtics mannitol or acetazolamide on nephroroxicity andphysiological disposition of cisplatin in rats. Cancer Che-mother Pharmacol 1358-6 2. 1984I5 I. McGinness JE, Proctor PH , D emopoulos HB:Ame-liontion of cis-platinum nephrotoxicity by orgotein (suprox-ide dismu tase). Physiol Chem Phys 10:267-277. 1978152. Beny JP. Pauwells C. l lw z c au S. et al: Effect of sele-nium in comb ination with cis-dichlorodiammineplatinum u) inthe treatment of murine fibrosarcoma. Cancer Res 44:2864-2868, 1984153. Campbell B. Kalrnan SM, Jacobs C: Plasma platinumlevels: Relationship to cisplatin do se and nephrotoxicity. Can-cer Treat Rep 67: 169-172. 1983154. Mihich G. Bullard G. Pavelic Z, et al: Preclinical stud-ies of dihydroxycisdichlorc+bis-isopropylarnineplatinum IV(CHIP). Proc Am Soc Clin Oncol 20:426. 1979 (abstr C-559 )155. Atassi G. Rozencweig M: Cisdichloro I . I . d i ( a m i n emethyl)cyclohexane platinum (II) (DD CP: NSC 26%08): Anew platinum complex with no renal toxicity. Proc Am SocClin Oncol 21:260. 1980 (abstr 1 044)156. Creaven PJ. Madajewicz S. Pendyala C. et al: Phase Ic l in ica l t r i a l of cis-dichloro-trans-dihydroxy-bis-isopropylamine platinum (IV ) (CH IP). Cancer Treat Rep67:795-800. 1983

    157. Rose WC. Schurig JE. Huftalen JB. et al: Antitumoractivity and toxicity of cisplatin analogs. Cancer Treat Rep66: 135- 146, 1982158. Rozencweiq M. Nicaise C. Beer M. et al: Phase I studyof carboplatin given on a five day intravenous schedule. J ClinOncol 1 :62 1-626, 1983


Recommended