Preventing and Managing Toxicities of High-Dose Methotrexate

Scott C Howard, John McCormick, Ching-Hon Pui, Randall K Buddington, R Donald Harvey, Scott C Howard, John McCormick, Ching-Hon Pui, Randall K Buddington, R Donald Harvey

Abstract

: High-dose methotrexate (HDMTX), defined as a dose higher than 500 mg/m2, is used to treat a range of adult and childhood cancers. Although HDMTX is safely administered to most patients, it can cause significant toxicity, including acute kidney injury (AKI) in 2%-12% of patients. Nephrotoxicity results from crystallization of methotrexate in the renal tubular lumen, leading to tubular toxicity. AKI and other toxicities of high-dose methotrexate can lead to significant morbidity, treatment delays, and diminished renal function. Risk factors for methotrexate-associated toxicity include a history of renal dysfunction, volume depletion, acidic urine, and drug interactions. Renal toxicity leads to impaired methotrexate clearance and prolonged exposure to toxic concentrations, which further worsen renal function and exacerbate nonrenal adverse events, including myelosuppression, mucositis, dermatologic toxicity, and hepatotoxicity. Serum creatinine, urine output, and serum methotrexate concentration are monitored to assess renal clearance, with concurrent hydration, urinary alkalinization, and leucovorin rescue to prevent and mitigate AKI and subsequent toxicity. When delayed methotrexate excretion or AKI occurs despite preventive strategies, increased hydration, high-dose leucovorin, and glucarpidase are usually sufficient to allow renal recovery without the need for dialysis. Prompt recognition and effective treatment of AKI and associated toxicities mitigate further toxicity, facilitate renal recovery, and permit patients to receive other chemotherapy or resume HDMTX therapy when additional courses are indicated.

Implications for practice: High-dose methotrexate (HDMTX), defined as a dose higher than 500 mg/m2, is used for a range of cancers. Although HDMTX is safely administered to most patients, it can cause significant toxicity, including acute kidney injury (AKI), attributable to crystallization of methotrexate in the renal tubular lumen, leading to tubular toxicity. When AKI occurs despite preventive strategies, increased hydration, high-dose leucovorin, and glucarpidase allow renal recovery without the need for dialysis. This article, based on a review of the current associated literature, provides comprehensive recommendations for prevention of toxicity and, when necessary, detailed treatment guidance to mitigate AKI and subsequent toxicity.

Keywords: Acute kidney injury; Glucarpidase; High-dose methotrexate; Leucovorin; Methotrexate; Pharmacokinetics.

Conflict of interest statement

of potential conflicts of interest may be found at the end of this article.

©AlphaMed Press.

Figures

Figure 1.
Figure 1.
Mechanism and site of action of MTX and of rescue strategies for delayed MTX elimination. After MTX enters cells through the RFC, it is polyglutamated, then competitively and reversibly inhibits the activity of DHFR, thus preventing formation of FH4 from FH2. The lack of FH4 inhibits DNA, RNA, and protein synthesis. LV enters cells through the RFC and allows formation of FH4 despite the presence of MTX, which effectively rescues cells. However, when MTX elimination is impaired and it is present at very high concentrations, very high doses of LV are necessary to allow entry of a sufficient amount to rescue cells from MTX toxicity. Glucarpidase eliminates extracellular MTX by converting it to nontoxic DAMPA and therefore should always be given with LV to provide intracellular rescue even as the glucarpidase prevents further accumulation of intracellular MTX by removing it from the extracellular compartment. Abbreviations: DAMPA, 4-deoxy-4-amino-N-10-methylpteroic acid; DHFR, dihydrofolate reductase; dUMP, deoxyuridine monophosphate; FH2, dihydrofolate; FH4, tetrahydrofolate; FH5, tetrahydrofolate; LV, leucovorin; MTX, methotrexate; RFC, reduced folate carrier; TMP, thymidine monophosphate.
Figure 2.
Figure 2.
Nomogram for the expected time-dependent decrease in serum MTX levels after completion of MTX infusion. Nomogram for expected serum MTX levels as a function of time from the end of a 6-hour infusion of methotrexate at a dose of 7.5 g/m2. The dark blue area represents ±2 SD from the mean (orange line). Values above the red line indicate impending or severe toxicity. Adapted from [112] with permission. Abbreviation: MTX, methotrexate.
Figure 3.
Figure 3.
Pharmacokinetically guided leucovorin rescue based on plasma MTX levels after high-dose MTX. Leucovorin dosing must be increased dramatically when plasma MTX levels are elevated above 5 µM at 42 hours after the start of the MTX infusion because leucovorin must compete with MTX to enter cells via the reduced folate carrier and the goal of leucovorin rescue is to achieve a high intracellular concentration of leucovorin. In color are the recommended doses of leucovorin based on the plasma MTX concentration at each time point after the start of the MTX infusion. For example, if at hour 60 the MTX concentration is 100 µM, it falls above the red line and the recommended leucovorin dose would be 1,000 mg/m2 every 6 hours. If at 100 hours the methotrexate concentration decreases to 3 µM (above the yellow line, below the orange line), then the recommended leucovorin dose would decrease to 10 mg/m2 every 3 hours. The dotted lines indicate extrapolated values based on modeling and clinical trial experience following the original publication [113]. Abbreviation: MTX, methotrexate.

References

    1. Widemann BC, Adamson PC. Understanding and managing methotrexate nephrotoxicity. The Oncologist. 2006;11:694–703.
    1. Stoller RG, Hande KR, Jacobs SA, et al. Use of plasma pharmacokinetics to predict and prevent methotrexate toxicity. N Engl J Med. 1977;297:630–634.
    1. Mitchell MS, Wawro NW, DeConti RC, et al. Effectiveness of high-dose infusions of methotrexate followed by leucovorin in carcinoma of the head and neck. Cancer Res. 1968;28:1088–1094.
    1. Abrey LE, DeAngelis LM, Yahalom J. Long-term survival in primary CNS lymphoma. J Clin Oncol. 1998;16:859–863.
    1. Christensen AM, Pauley JL, Molinelli AR, et al. Resumption of high-dose methotrexate after acute kidney injury and glucarpidase use in pediatric oncology patients. Cancer. 2012;118:4321–4330.
    1. Widemann BC, Balis FM, Kim A, et al. Glucarpidase, leucovorin, and thymidine for high-dose methotrexate-induced renal dysfunction: Clinical and pharmacologic factors affecting outcome. J Clin Oncol. 2010;28:3979–3986.
    1. Perazella MA, Moeckel GW. Nephrotoxicity from chemotherapeutic agents: Clinical manifestations, pathobiology, and prevention/therapy. Semin Nephrol. 2010;30:570–581.
    1. May J, Carson KR, Butler S, et al. High incidence of methotrexate associated renal toxicity in patients with lymphoma: A retrospective analysis. Leuk Lymphoma. 2014;55:1345–1349.
    1. de Miguel D, García-Suárez J, Martín Y, et al. Severe acute renal failure following high-dose methotrexate therapy in adults with haematological malignancies: A significant number result from unrecognized co-administration of several drugs. Nephrol Dial Transplant. 2008;23:3762–3766.
    1. Suzuki K, Doki K, Homma M, et al. Co-administration of proton pump inhibitors delays elimination of plasma methotrexate in high-dose methotrexate therapy. Br J Clin Pharmacol. 2009;67:44–49.
    1. Dao K, Ivanyuk A, Buclin T, et al. Pharmacokinetic interaction between methotrexate and chloral hydrate. Pediatr Blood Cancer. 2013;60:518–520.
    1. Bain E, Birhiray RE, Reeves DJ. Drug-drug interaction between methotrexate and levetiracetam resulting in delayed methotrexate elimination. Ann Pharmacother. 2014;48:292–296.
    1. Parentelli AS, Phulpin-Weibel A, Mansuy L, et al. Drug-drug interaction between methotrexate and levetiracetam in a child treated for acute lymphoblastic leukemia. Pediatr Blood Cancer. 2013;60:340–341.
    1. McBride A, Antonia SJ, Haura EB, et al. Suspected methotrexate toxicity from omeprazole: a case review of carboxypeptidase G2 use in a methotrexate-experienced patient with methotrexate toxicity and a review of the literature. J Pharm Pract. 2012;25:477–485.
    1. Bezabeh S, Mackey AC, Kluetz P, et al. Accumulating evidence for a drug-drug interaction between methotrexate and proton pump inhibitors. The Oncologist. 2012;17:550–554.
    1. Meyers PA, Flombaum C. High-dose methotrexate-induced renal dysfunction: is glucarpidase necessary for rescue? J Clin Oncol. 2011;29:e180–e180; author reply e181.
    1. Basch E, Prestrud AA, Hesketh PJ, et al. Antiemetics: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2011;29:4189–4198.
    1. Ackland SP, Schilsky RL. High-dose methotrexate: A critical reappraisal. J Clin Oncol. 1987;5:2017–2031.
    1. Bhojwani D, Sabin ND, Pei D, et al. Methotrexate-induced neurotoxicity and leukoencephalopathy in childhood acute lymphoblastic leukemia. J Clin Oncol. 2014;32:949–959.
    1. Taylor SR, Banker A, Schlaen A, et al. Intraocular methotrexate can induce extended remission in some patients in noninfectious uveitis. Retina. 2013;33:2149–2154.
    1. D’Elia T. Methotrexate-induced pneumonitis: Heterogeneity of bronchoalveolar lavage and differences between cancer and rheumatoid arthritis. Inflamm Allergy Drug Targets. 2014;13:25–33.
    1. Jakubovic BD, Donovan A, Webster PM, et al. Methotrexate-induced pulmonary toxicity. Can Respir J. 2013;20:153–155.
    1. Sathi N, Chikura B, Kaushik VV, et al. How common is methotrexate pneumonitis? A large prospective study investigates. Clin Rheumatol. 2012;31:79–83.
    1. Elmorsi YM, El-Haggar SM, Ibrahim OM, et al. Effect of ketoprofen and indomethacin on methotrexate pharmacokinetics in mice plasma and tumor tissues. Eur J Drug Metab Pharmacokinet. 2013;38:27–32.
    1. Hardwick RN, Clarke JD, Lake AD, et al. Increased susceptibility to methotrexate-induced toxicity in nonalcoholic steatohepatitis. Toxicol Sci. 2014;142:45–55.
    1. Ekstrøm PO, Anderson A, Warren DJ, et al. Pharmacokinetics of different doses of methotrexate at steady state by in situ microdialysis in a rat model. Cancer Chemother Pharmacol. 1995;36:283–289.
    1. Fuskevåg O-M, Kristiansen C, Lindal S, et al. Leucovorin and maximum tolerated dose toxicity of methotrexate in rats. Pediatr Hematol Oncol. 2000;17:651–658.
    1. Asvadi I, Hajipour B, Asvadi A, et al. Protective effect of pentoxyfilline in renal toxicity after methotrexate administration. Eur Rev Med Pharmacol Sci. 2011;15:1003–1009.
    1. Chen C, Tian L, Zhang M, et al. Protective effect of amifostine on high-dose methotrexate-induced small intestinal mucositis in mice. Dig Dis Sci. 2013;58:3134–3143.
    1. Abraham P, Kolli VK, Rabi S. Melatonin attenuates methotrexate-induced oxidative stress and renal damage in rats. Cell Biochem Funct. 2010;28:426–433.
    1. Ibrahim MA, El-Sheikh AA, Khalaf HM, et al. Protective effect of peroxisome proliferator activator receptor (PPAR)-α and -γ ligands against methotrexate-induced nephrotoxicity. Immunopharmacol Immunotoxicol. 2014;36:130–137.
    1. Jacobs SA, Stoller RG, Chabner BA, et al. 7-Hydroxymethotrexate as a urinary metabolite in human subjects and rhesus monkeys receiving high dose methotrexate. J Clin Invest. 1976;57:534–538.
    1. Chabner BA, Johns DG, Bertino JR. Enzymatic cleavage of methotrexate provides a method for prevention of drug toxicity. Nature. 1972;239:395–397.
    1. Garnick M, Mayer R, Abelson H. Acute renal failure associated with cancer treatment. In: Brenner BM, Lazarus JM, eds. Acute Renal Failure. New York: Churchill Livingstone, 1988:621–657
    1. Pitman S, Parker L, Tattersall M, et al. Clinical trial of high-dose methotrexate (nsc-740) with citrovorum factor (nsc-3590)-toxicologic and therapeutic observations. Cancer Chemother Rep. 1975;6:43–49.
    1. Pitman SW, Frei E., 3rd Weekly methotrexate-calcium leucovorin rescue: effect of alkalinization on nephrotoxicity; pharmacokinetics in the CNS; and use in CNS non-Hodgkin’s lymphoma. Cancer Treat Rep. 1977;61:695–701.
    1. Lau KK, Weiss AR, Jones DP. Polyuria associated with high-dose methotrexate in two patients with acute lymphoblastic leukaemia. J Oncol Pharm Pract. 2005;11:31–33.
    1. Sahni V, Choudhury D, Ahmed Z. Chemotherapy-associated renal dysfunction. Nat Rev Nephrol. 2009;5:450–462.
    1. Xu W, Zhang L-y, Chen X-y, et al. Serum creatinine and creatinine clearance for predicting plasma methotrexate concentrations after high-dose methotrexate chemotherapy for the treatment for childhood lymphoblastic malignancies. Cancer Chemother Pharmacol. 2014;73:79–86.
    1. Kintzel PE, Dorr RT. Anticancer drug renal toxicity and elimination: Dosing guidelines for altered renal function. Cancer Treat Rev. 1995;21:33–64.
    1. Aronoff GR, Berns J, Brier M, et al. Drug prescribing in renal failure:Dosing guidelines for adults. 1999448.
    1. Chiusolo P, Giammarco S, Bellesi S, et al. The role of MTHFR and RFC1 polymorphisms on toxicity and outcome of adult patients with hematological malignancies treated with high-dose methotrexate followed by leucovorin rescue. Cancer Chemother Pharmacol. 2012;69:691–696.
    1. Treviño LR, Shimasaki N, Yang W, et al. Germline genetic variation in an organic anion transporter polypeptide associated with methotrexate pharmacokinetics and clinical effects. J Clin Oncol. 2009;27:5972–5978.
    1. Ramsey LB, Bruun GH, Yang W, et al. Rare versus common variants in pharmacogenetics: SLCO1B1 variation and methotrexate disposition. Genome Res. 2012;22:1–8.
    1. Ramsey LB, Panetta JC, Smith C, et al. Genome-wide study of methotrexate clearance replicates SLCO1B1. Blood. 2013;121:898–904.
    1. Wright KD, Panetta JC, Onar-Thomas A, et al. Delayed methotrexate excretion in infants and young children with primary central nervous system tumors and postoperative fluid collections. Cancer Chemother Pharmacol. 2015;75:27–35.
    1. Gregory RE, Pui CH, Crom WR. Raised plasma methotrexate concentrations following intrathecal administration in children with renal dysfunction. Leukemia. 1991;5:999–1003.
    1. Kelly H, Harvey D, Moll S. A cautionary tale: fatal outcome of methotrexate therapy given for management of ectopic pregnancy. Obstet Gynecol. 2006;107:439–441.
    1. Chen S. Retooling the creatinine clearance equation to estimate kinetic GFR when the plasma creatinine is changing acutely. J Am Soc Nephrol. 2013;24:877–888.
    1. Widemann BC, Schwartz S, Jayaprakash N, et al. Efficacy of glucarpidase (carboxypeptidase g2) in patients with acute kidney injury after high‐dose methotrexate therapy. Pharmacotherapy. 2014;34:427–439.
    1. Relling MV, Fairclough D, Ayers D, et al. Patient characteristics associated with high-risk methotrexate concentrations and toxicity. J Clin Oncol. 1994;12:1667–1672.
    1. Shamash J, Earl H, Souhami R. Acetazolamide for alkalinisation of urine in patients receiving high-dose methotrexate. Cancer Chemother Pharmacol. 1991;28:150–151.
    1. Evans WE, Pratt CB, Taylor RH, et al. Pharmacokinetic monitoring of high-dose methotrexate. Early recognition of high-risk patients. Cancer Chemother Pharmacol. 1979;3:161–166.
    1. Al-Turkmani MR, Law T, Narla A, et al. Difficulty measuring methotrexate in a patient with high-dose methotrexate-induced nephrotoxicity. Clin Chem. 2010;56:1792–1794.
    1. Ylinen E, Jahnukainen K, Saarinen-Pihkala UM, et al. Assessment of renal function during high-dose methotrexate treatment in children with acute lymphoblastic leukemia. Pediatr Blood Cancer. 2014;61:2199–2202.
    1. Aumente D, Buelga DS, Lukas JC, et al. Population pharmacokinetics of high-dose methotrexate in children with acute lymphoblastic leukaemia. Clin Pharmacokinet. 2006;45:1227–1238.
    1. Plard C, Bressolle F, Fakhoury M, et al. A limited sampling strategy to estimate individual pharmacokinetic parameters of methotrexate in children with acute lymphoblastic leukemia. Cancer Chemother Pharmacol. 2007;60:609–620.
    1. Pui C-H, Sandlund JT, Pei D, et al. Improved outcome for children with acute lymphoblastic leukemia: Results of Total Therapy Study XIIIB at St Jude Children’s Research Hospital. Blood. 2004;104:2690–2696.
    1. Tsurusawa M, Gosho M, Mori T, et al. Statistical analysis of relation between plasma methotrexate concentration and toxicity in high-dose methotrexate therapy of childhood nonHodgkin lymphoma. Pediatr Blood Cancer. 2014 [Epub ahead of print]
    1. Howard SC, Pedrosa M, Lins M, et al. Establishment of a pediatric oncology program and outcomes of childhood acute lymphoblastic leukemia in a resource-poor area. JAMA. 2004;291:2471–2475.
    1. Maia MB, Saivin S, Chatelut E, et al. In vitro and in vivo protein binding of methotrexate assessed by microdialysis. Int J Clin Pharmacol Ther. 1996;34:335–341.
    1. Saland JM, Leavey PJ, Bash RO, et al. Effective removal of methotrexate by high-flux hemodialysis. Pediatr Nephrol. 2002;17:825–829.
    1. Wall SM, Johansen MJ, Molony DA, et al. Effective clearance of methotrexate using high-flux hemodialysis membranes. Am J Kidney Dis. 1996;28:846–854.
    1. Widemann BC, Balis FM, Murphy RF, et al. Carboxypeptidase-G2, thymidine, and leucovorin rescue in cancer patients with methotrexate-induced renal dysfunction. J Clin Oncol. 1997;15:2125–2134.
    1. Relling MV, Stapleton FB, Ochs J, et al. Removal of methotrexate, leucovorin, and their metabolites by combined hemodialysis and hemoperfusion. Cancer. 1988;62:884–888.
    1. Lindsay K, Fraser AD, Layton A, et al. Liver fibrosis in patients with psoriasis and psoriatic arthritis on long-term, high cumulative dose methotrexate therapy. Rheumatology (Oxford) 2009;48:569–572.
    1. Weber BL, Tanyer G, Poplack DG, et al. Transient acute hepatotoxicity of high-dose methotrexate therapy during childhood NCI Monogr 1987207–212.
    1. Locasciulli A, Mura R, Fraschini D, et al. High-dose methotrexate administration and acute liver damage in children treated for acute lymphoblastic leukemia. A prospective study. Haematologica. 1992;77:49–53.
    1. Nyfors A. Liver biopsies from psoriatics related to methotrexate therapy. 3. Findings in post-methotrexate liver biopsies from 160 psoriatics. Acta Pathol Microbiol Scand [A] 1977;85:511–518.
    1. Watson WA, Litovitz TL, Rodgers GC, Jr, et al. 2004 annual report of the American Association of Poison Control Centers toxic exposure surveillance system. Am J Emerg Med. 2005;23:589–666.
    1. Kremer JM, Alarcón GS, Lightfoot RW, Jr, et al. Methotrexate for rheumatoid arthritis. Suggested guidelines for monitoring liver toxicity. Arthritis Rheum. 1994;37:316–328.
    1. Mahoney DH, Jr, Shuster JJ, Nitschke R, et al. Acute neurotoxicity in children with B-precursor acute lymphoid leukemia: An association with intermediate-dose intravenous methotrexate and intrathecal triple therapy--a Pediatric Oncology Group study. J Clin Oncol. 1998;16:1712–1722.
    1. Rubnitz JE, Relling MV, Harrison PL, et al. Transient encephalopathy following high-dose methotrexate treatment in childhood acute lymphoblastic leukemia. Leukemia. 1998;12:1176–1181.
    1. Cronstein BN, Naime D, Ostad E. The antiinflammatory mechanism of methotrexate. Increased adenosine release at inflamed sites diminishes leukocyte accumulation in an in vivo model of inflammation. J Clin Invest. 1993;92:2675–2682.
    1. Bernini JC, Fort DW, Griener JC, et al. Aminophylline for methotrexate-induced neurotoxicity. Lancet. 1995;345:544–547.
    1. Sonis ST. Mucositis as a biological process: A new hypothesis for the development of chemotherapy-induced stomatotoxicity. Oral Oncol. 1998;34:39–43.
    1. Sonis ST. The pathobiology of mucositis. Nat Rev Cancer. 2004;4:277–284.
    1. Sonis ST. A biological approach to mucositis. J Support Oncol. 2004;2:21–32; discussion 35–36.
    1. Dumontet C, Sonnet A, Bastion Y, et al. Prevention of high dose L-PAM-induced mucositis by cryotherapy. Bone Marrow Transplant. 1994;14:492–494.
    1. Edelman MJ, Gandara DR, Perez EA, et al. Phase I trial of edatrexate plus carboplatin in advanced solid tumors: Amelioration of dose-limiting mucositis by ice chip cryotherapy. Invest New Drugs. 1998;16:69–75.
    1. Matejka M, Nell A, Kment G, et al. Local benefit of prostaglandin E2 in radiochemotherapy-induced oral mucositis. Br J Oral Maxillofac Surg. 1990;28:89–91.
    1. Schmidt E, Thoennissen NH, Rudat A, et al. Use of palifermin for the prevention of high-dose methotrexate-induced oral mucositis. Ann Oncol. 2008;19:1644–1649.
    1. Maiguma T, Kaji H, Makino K, et al. Protective effects of amifostine and cyclooxygenase-1 inhibitor against normal human epidermal keratinocyte toxicity induced by methotrexate and 5-fluorouracil. Basic Clin Pharmacol Toxicol. 2009;105:1–9.
    1. Worthington HV, Clarkson JE, Bryan G, et al. Interventions for preventing oral mucositis for patients with cancer receiving treatment. Cochrane Database Syst Rev. 2011;4:CD000978.
    1. Clarkson JE, Worthington HV, Furness S, et al. Interventions for treating oral mucositis for patients with cancer receiving treatment. Cochrane Database Syst Rev. 2010;8:CD001973.
    1. Sukhotnik I, Pollak Y, Coran AG, et al. Glutamine attenuates the inhibitory effect of methotrexate on TLR signaling during intestinal chemotherapy-induced mucositis in a rat. Nutr Metab (Lond) 2014;11:17.
    1. Hamada K, Kakigawa N, Sekine S, et al. Disruption of ZO-1/claudin-4 interaction in relation to inflammatory responses in methotrexate-induced intestinal mucositis. Cancer Chemother Pharmacol. 2013;72:757–765.
    1. Fijlstra M, Tissing WJ, Stellaard F, et al. Reduced absorption of long-chain fatty acids during methotrexate-induced gastrointestinal mucositis in the rat. Clin Nutr. 2013;32:452–459.
    1. Goh TS, Wong KY, Lampkin B, et al. Evaluation of 24-hour infusion of high-dose methotrexate--pharmacokinetics and toxicity. Cancer Chemother Pharmacol. 1979;3:177–180.
    1. Widemann BC, Hetherington ML, Murphy RF, et al. Carboxypeptidase-G2 rescue in a patient with high dose methotrexate-induced nephrotoxicity. Cancer. 1995;76:521–526.
    1. Pinedo HM, Zaharko DS, Bull JM, et al. The reversal of methotrexate cytotoxicity to mouse bone marrow cells by leucovorin and nucleosides. Cancer Res. 1976;36:4418–4424.
    1. Mulder RL, Knijnenburg SL, Geskus RB, et al. Glomerular function time trends in long-term survivors of childhood cancer: A longitudinal study. Cancer Epidemiol Biomarkers Prev. 2013;22:1736–1746.
    1. Schwartz S, Borner K, Müller K, et al. Glucarpidase (carboxypeptidase g2) intervention in adult and elderly cancer patients with renal dysfunction and delayed methotrexate elimination after high-dose methotrexate therapy. The Oncologist. 2007;12:1299–1308.
    1. Widemann BC, Jayaprakash N, Howard SC, et al. Clinical trial and compassionate use experience with glucarpidase for methotrexate toxicity. J Clin Oncol. 2012;30(suppl):6530.
    1. Buchen S, Ngampolo D, Melton RG, et al. Carboxypeptidase G2 rescue in patients with methotrexate intoxication and renal failure. Br J Cancer. 2005;92:480–487.
    1. Martin J, Howard SC, Pillai A, et al. The weaned pig as a model for Doxorubicin-induced mucositis. Chemotherapy. 2014;60:24–36.
    1. Wang J, Li G. Mechanisms of methotrexate resistance in osteosarcoma cell lines and strategies for overcoming this resistance. Oncol Lett. 2015;9:940–944.
    1. Takeuchi J, Kyo T, Naito K, et al. Induction therapy by frequent administration of doxorubicin with four other drugs, followed by intensive consolidation and maintenance therapy for adult acute lymphoblastic leukemia: The JALSG-ALL93 study. Leukemia. 2002;16:1259–1266.
    1. Linker C, Damon L, Ries C, et al. Intensified and shortened cyclical chemotherapy for adult acute lymphoblastic leukemia. J Clin Oncol. 2002;20:2464–2471.
    1. Hill FG, Richards S, Gibson B, et al. Successful treatment without cranial radiotherapy of children receiving intensified chemotherapy for acute lymphoblastic leukaemia: Results of the risk-stratified randomized central nervous system treatment trial MRC UKALL XI (ISRC TN 16757172) Br J Haematol. 2004;124:33–46.
    1. Zhang HN, He XL, Wang C, et al. Impact of SLCO1B1 521T>C variant on leucovorin rescue and risk of relapse in childhood acute lymphoblastic leukemia treated with high-dose methotrexate. Pediatr Blood Cancer. 2014;61:2203–2207.
    1. Souhami RL, Craft AW, Van der Eijken JW, et al. Randomised trial of two regimens of chemotherapy in operable osteosarcoma: A study of the European Osteosarcoma Intergroup. Lancet. 1997;350:911–917.
    1. Fuchs N, Bielack SS, Epler D, et al. Long-term results of the co-operative German-Austrian-Swiss osteosarcoma study group’s protocol COSS-86 of intensive multidrug chemotherapy and surgery for osteosarcoma of the limbs. Ann Oncol. 1998;9:893–899.
    1. Bacci G, Briccoli A, Ferrari S, et al. Neoadjuvant chemotherapy for osteosarcoma of the extremity: Long-term results of the Rizzoli’s 4th protocol. Eur J Cancer. 2001;37:2030–2039.
    1. Goorin AM, Schwartzentruber DJ, Devidas M, et al. Presurgical chemotherapy compared with immediate surgery and adjuvant chemotherapy for nonmetastatic osteosarcoma: Pediatric Oncology Group Study POG-8651. J Clin Oncol. 2003;21:1574–1580.
    1. Ferrari S, Smeland S, Mercuri M, et al. Neoadjuvant chemotherapy with high-dose Ifosfamide, high-dose methotrexate, cisplatin, and doxorubicin for patients with localized osteosarcoma of the extremity: A joint study by the Italian and Scandinavian Sarcoma Groups. J Clin Oncol. 2005;23:8845–8852.
    1. Koller CA, Kantarjian HM, Thomas D, et al. The hyper-CVAD regimen improves outcome in relapsed acute lymphoblastic leukemia. Leukemia. 1997;11:2039–2044.
    1. Khouri IF, Romaguera J, Kantarjian H, et al. Hyper-CVAD and high-dose methotrexate/cytarabine followed by stem-cell transplantation: An active regimen for aggressive mantle-cell lymphoma. J Clin Oncol. 1998;16:3803–3809.
    1. Thomas DA, O’Brien S, Cortes J, et al. Outcome with the hyper-CVAD regimens in lymphoblastic lymphoma. Blood. 2004;104:1624–1630.
    1. Batchelor T, Carson K, O’Neill A, et al. Treatment of primary CNS lymphoma with methotrexate and deferred radiotherapy: A report of NABTT 96-07. J Clin Oncol. 2003;21:1044–1049.
    1. Dalia S, Price S, Forsyth P, et al. What is the optimal dose of high-dose methotrexate in the initial treatment of primary central nervous system lymphoma? Leuk Lymphoma. 2015;56:500–502.
    1. Abelson HT, Fosburg MT, Beardsley GP, et al. Methotrexate-induced renal impairment: Clinical studies and rescue from systemic toxicity with high-dose leucovorin and thymidine. J Clin Oncol. 1983;1:208–216.
    1. Bleyer WA. Methotrexate: Clinical pharmacology, current status and therapeutic guidelines. Cancer Treat Rev. 1977;4:87–101.

Source: PubMed

3
Sottoscrivi