Elsevier

The Lancet Oncology

Volume 11, Issue 11, November 2010, Pages 1096-1106
The Lancet Oncology

Review
Glucocorticoid use in acute lymphoblastic leukaemia

https://doi.org/10.1016/S1470-2045(10)70114-5Get rights and content

Summary

Glucocorticoids (prednisone and dexamethasone) play an essential part in the treatment of acute lymphoblastic leukaemia (ALL), but their optimum doses and bioequivalence have not been established. Results of preclinical studies have shown that dexamethasone has a longer half-life and better CNS penetration than does prednisone. In prospective randomised trials, dexamethasone improved control of CNS leukaemia. At a prednisone-to-dexamethasone dose ratio of less than seven, dexamethasone (6–18 mg/m2 per day) resulted in a better event-free survival than did prednisone (40–120 mg/m2 per day), and high-dose dexamethasone (10–18 mg/m2 per day) improved the outcome of T-cell ALL and high-risk ALL. However, dexamethasone caused more adverse effects, including infection, bone fracture, osteonecrosis, mood and behaviour problems, and myopathy. At a dose ratio greater than seven, the two drugs showed no difference in efficacy. Therefore, the efficacy of prednisone and dexamethasone is dose dependent and needs to be carefully assessed against the toxic effects. Moreover, although dexamethasone generally showed increased activity in ALL cells in vitro, the dose ratio of the two drugs that exerted equivalent cytotoxic effects differed substantially in samples from individuals. The selection of the type and dose of glucocorticoid should be based on the risk of relapse, treatment phase, and the chemotherapeutic drugs used concomitantly.

Introduction

Glucocorticoids were among the first drug classes used in the treatment of patients with acute lymphoblastic leukaemia (ALL) and are still essential components of treament.1, 2 They seem to exert their cytotoxic effects by binding to glucocorticoid receptors in the cytoplasm (figure 1).3 These receptors can then form dimers, translocate to the nucleus, and interact with glucocorticoid-response elements to transactivate gene expression, or they can remain as monomers and repress the activity of transcription factors such as the activating protein-1 (AP-1) or nuclear factor-κB (NFκB).4, 5, 6 Both pathways inhibit cytokine production,7 change the expression of various oncogenes,8 and induce cell-cycle arrest9 and apoptosis.10

In vivo and in vitro, glucocorticoid resistance is an adverse prognostic factor in ALL, and several mechanisms have been reported.1 Glucocorticoid exposure induces upregulation of the glucocorticoid receptor in ALL cells, and about half of 51 responsive genes identified have been functionally linked to three major pathways: cell proliferation and survival (mitogen-activated protein kinase [MAPK] pathways), NFκB signalling, and glucose metabolism.11, 12 Glucocorticoid resistance has been associated with upregulation of the genes involved in glucose metabolism, and increased glucose uptake into the cells.11, 13, 14 Glucocorticoids also induce release of calcium ions from the endoplasmic reticulum into the cytosol; the resulting increase in mitochondrial calcium ions induces cytochrome-c release and triggers apoptosis. Raised expression of the calcium-binding proteins (CBP) S100A8 and S100A9, and of the antiapoptotic B-cell lymphoma 2 (BCL-2) protein family member myeloid cell leukaemia sequence 1 (MCL-1), inhibit signalling mediated by free cytosolic and mitochondrial calcium ions signals, respectively, causing glucocorticoid resistance.15, 16, 17

Traditionally, prednisone has been the glucocorticoid most commonly used in the treatment of patients with ALL; it is typically given for 4 consecutive weeks in combination with vincristine, an anthracycline, asparaginase, and intrathecal chemotherapy. In the past few years, dexamethasone, another glucocorticoid, has been used increasingly to treat ALL. These two glucocorticoids are synthetic analogues of cortisol that differ molecularly in several important aspects (figure 2).18, 19, 20, 21 Dexamethasone differs from prednisolone (active metabolite of prednisone) only by a fluorine atom at the 9α position of ring B and a methyl group at the C16 position of ring D. The 9α fluorine slows the metabolism of dexamethasone, thereby extending its plasma half-life (200 min vs 60 min for prednisolone) and biological half-life (36–54 h vs 24–36 h).19, 21 The C16 methyl group reduces the sodium-retention effect of dexamethasone to a minimum. Prednisone is thought to have half the mineral corticoid activity of cortisol, whereas dexamethasone is thought to have little or none.21

Results from bioequivalence studies of dexamethasone and prednisone have often been discordant. Generally, 1 mg of dexamethasone is equivalent to 5–10 mg of prednisone in reducing inflammation.20, 21, 22 However, this equivalence has not been firmly established in antileukaemic treatment. We review the use of prednisone and dexamethasone in the treatment of ALL, assessing evidence from in-vitro studies, preclinical models, and clinical studies; comparing the benefits and adverse effects of the drugs, and discussing their optimum uses.

Section snippets

In-vitro cytotoxic effects

The cytotoxic effects of prednisolone and dexamethasone were compared in samples of ALL cells from 133 untreated children by use of the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in cell-suspension cultures.23 The cytotoxic effects of dexamethasone were much greater than those of prednisolone, and were much greater than those predicted according to the relative anti-inflammatory effects of the drugs. The median concentration that produces 50% cytotoxicity (LC50)

CNS penetration of glucocorticoids

The success of treatment of ALL depends on effective CNS-directed therapy, and with the reduced use and eventual elimination of prophylactic cranial irradiation, glucocorticoids play an increasingly important part in the control of CNS leukaemia.29 Glucocorticoids are used to treat a range of neurological disorders, such as cerebral oedema, haemorrhage, meningitis, and epilepsy.30 Dexamethasone is the preferred glucocorticoid for these conditions because of its long half-life, excellent CNS

Early clinical trials

The 6·67 dose ratio of prednisone (prednisolone) to dexamethasone used in a non-human primate model was tested in early clinical trials (table).31 In 1971, the Cancer and Leukaemia Group B (CALGB) did the first randomised trial of 493 children with ALL (table).33 Patients were given either prednisone 40 mg/m2 per day or dexamethasone 6 mg/m2 per day during remission-induction and maintenance treatment. Dexamethasone significantly reduced the frequency of CNS relapse compared with prednisone in

Randomised clinical trials

The DFCI 91-01 protocol included a 3-day treatment phase in which 369 children with ALL were randomly assigned to prednisolone (40 mg/m2 per day) or dexamethasone (6 mg/m2 per day, 18 mg/m2 per day, or 150 mg/m2 per day) before remission-induction treatment (table).36, 39 The dose of dexamethasone was positively associated with the degree of bone-marrow response. High-dose dexamethasone abrogated the effect of drug insensitivity and of low glucocorticoid-receptor expression on leukaemia blast

Adverse effects

Side-effects of glucocorticoids are dependent on the dose and the duration of treatment. The toxic effects of prednisone and dexamethasone have not been fully investigated in patients with ALL, but are most often reported with dexamethasone (table). Therefore, any benefits of dexamethasone must be assessed against the risk of toxic and adverse effects.

Long-term exposure to high-dose dexamethasone and myelosuppressive treatment can cause severe infections during remission-induction treatment and

Future studies

The remarkable progress in the treatment of childhood ALL warrants an increased effort to reduce the morbidity and toxic effects of glucocorticoids, without compromising their antileukaemic benefits. Because the LC50 and LC90 ratio of prednisolone to dexamethasone vary greatly in vitro23, 24 and the activities of the two drugs differ in vivo, a clinical trial could reasonably use both prednisone and dexamethasone during the 2–3 year treatment regimen for patients with ALL unless sensitivity to

Conclusion

Dexamethasone reduces the frequency of CNS relapse and is more effective than prednisone at a dose ratio of prednisone to dexamethasone below 7. When the dose ratio is greater than 7, EFS estimates are comparable with the two drugs, although dexamethasone still appears to yield improved CNS control. A high dose (eg, dexamethasone at 10 to 18 mg/m2 per day) can overcome drug resistance in T-cell ALL and high-risk ALL. The toxic effects associated with prednisone treatment are generally less than

Search strategy and selection criteria

We searched Medline and PubMed for articles published in English from Jan 1, 1950 to March 31, 2010, using the search terms “acute lymphoblastic leukemia,” “glucocorticoid,” “prednisone,” “prednisolone,” and “dexamethasone.” Additional information was obtained from abstracts presented at the American Society of Hematology annual meeting in 2003, 2008, and 2009.

References (76)

  • OC Green et al.

    Plasma levels, half-life values, and correlation with physiologic assays for growth and immunity

    J Pediatr

    (1978)
  • LB Silverman et al.

    Improved outcome for children with acute lymphoblastic leukemia: results of Dana-Farber Consortium Protocol 91-01

    Blood

    (2001)
  • BC Bostrom et al.

    Dexamethasone versus prednisone and daily oral versus weekly intravenous mercaptopurine for patients with standard-risk acute lymphoblastic leukemia: a report from the Children's Cancer Group

    Blood

    (2003)
  • NS Kadan-Lottick et al.

    A comparison of neurocognitive functioning in children previously randomized to dexamethasone or prednisone in the treatment of childhood acute lymphoblastic leukemia

    Blood

    (2009)
  • CH Pui et al.

    Early intensification of intrathecal chemotherapy virtually eliminates central nervous system relapse in children with acute lymphoblastic leukemia

    Blood

    (1998)
  • CH Pui 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)
  • OM Wolkowitz

    Prospective controlled studies of the behavioral and biological effects of exogenous corticosteroids

    Psychoneuroendocrinology

    (1994)
  • V Conter et al.

    Pulses of vincristine and dexamethasone in addition to intensive chemotherapy for children with intermediate-risk acute lymphoblastic leukaemia: a multicentre randomised trial

    Lancet

    (2007)
  • AJ Veerman et al.

    Dexamethasone-based therapy for childhood acute lymphoblastic leukaemia: results of the prospective Dutch Childhood Oncology Group (DCOG) protocol ALL-9 (1997–2004)

    Lancet Oncol

    (2009)
  • E Coustan-Smith et al.

    Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia

    Lancet Oncol

    (2009)
  • CH Pui

    T cell acute lymphoblastic leukemia: NOTCHing the way toward a better treatment outcome

    Cancer Cell

    (2009)
  • EM Te Poele et al.

    Dexamethasone in the maintenance phase of acute lymphoblastic leukaemia treatment: is the risk of lethal infections too high?

    Eur J Cancer

    (2007)
  • CH Pui et al.

    Treatment of acute lymphoblastic leukemia

    N Engl J Med

    (2006)
  • OH Pearson et al.

    Use of pituitary adrenocorticotropic hormone (ACTH) and cortisone in lymphomas and leukemias

    JAMA

    (1950)
  • FF Quddus et al.

    Glucocorticoid receptors in immunological subtypes of childhood acute lymphocytic leukemia cells: a Pediatric Oncology Group Study

    Cancer Res

    (1985)
  • WJ Tissing et al.

    Molecular determinants of glucocorticoid sensitivity and resistance in acute lymphoblastic leukemia

    Leukemia

    (2003)
  • A Ray et al.

    Physical association and functional antagonism between the p65 subunit of transcription factor NF-kappa B and the glucocorticoid receptor

    Proc Natl Acad Sci USA

    (1994)
  • SK Arya et al.

    Dexamethasone-mediated inhibition of human T cell growth factor and gamma-interferon messenger RNA

    J Immunol

    (1984)
  • M Schuchard et al.

    Steroid hormone regulation of nuclear proto-oncogenes

    Endocr Rev

    (1993)
  • JM Harmon et al.

    Dexamethasone induces irreversible G1 arrest and death of a human lymphoid cell line

    J Cell Physiol

    (1979)
  • AH Wyllie

    Glucocorticoid-induced thymocyte apoptosis is associated with endogenous endonuclease activation

    Nature

    (1980)
  • A Holleman et al.

    Gene-expression patterns in drug-resistant acute lymphoblastic leukemia cells and response to treatment

    N Engl J Med

    (2004)
  • Hagelstein J, Schneider P, de Boer J, et al. High expression of the Ca2+-binding proteins S100A8 and S100A9 cause...
  • HL Cantrill et al.

    In vitro determination of relative corticosteroid potency

    J Clin Endocrinol Metab

    (1975)
  • LI Rose et al.

    Choosing corticosteroid preparations

    Am Fam Physician

    (1978)
  • PL Ballard et al.

    A radioreceptor assay for evaluation of the plasma glucocorticoid activity of natural and synthetic steroids in man

    J Clin Endocrinol Metab

    (1975)
  • GJ Kaspers et al.

    Comparison of the antileukemic activity in vitro of dexamethasone and prednisolone in childhood acute lymphoblastic leukemia

    Med Pediatr Oncol

    (1996)
  • C Ito et al.

    Comparative cytotoxicity of dexamethasone and prednisolone in childhood acute lymphoblastic leukemia

    J Clin Oncol

    (1996)
  • Cited by (269)

    View all citing articles on Scopus
    View full text