Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 19.
Published in final edited form as: Eur J Neurol. 2009 Jun;16(6):653–655. doi: 10.1111/j.1468-1331.2009.02597.x

A proposed bailout for A-T patients?

Richard A Gatti a, Susan Perlman b
PMCID: PMC3242728  NIHMSID: NIHMS135267  PMID: 19459929

These are times of economic crises and proposed bailouts of failing systems. A proposal has recently been put forward for treating patients with ataxia-telangiectasia (A-T): Betamethasone partially restores neurological functions [13]. The questions being asked are very similar: Will it work? For how long? What are the potential dangers and costs? Most interestingly, there seem to be some unexpected lessons to be learned from clinical trials – and, thus, the metaphor may aptly apply back upon the economic sector as to which remedies work and which do not.

Ataxia-telangiectasia is an autosomal recessive cerebellar ataxia with onset between 1–4 years of age [47]. A-T arises from mutations in the ataxia-telangiectasia mutated (ATM) gene which encodes a ubiquitous serine-threonine kinase. The incidence of A-T is approximately one in 100 000 live births; however, this varies with the coefficient of inbreeding within an ethnic group. The most common presenting complaints are unsteadiness of gast and slurred speech. Apraxia of eye movements is almost always present as well. Cancer (e.g. leukemia, lymphoma, and others) affects roughly one-third of patients and is occasionally the presenting complaint. This creates a dangerous situation when radiation therapy is recommended without the realization that these patients are also radiosensitive and conventional doses of radiation are contraindicated and often lethal. A clinical diagnosis can be confirmed by elevated serum alphafetoprotein, an undetectable ATM protein on western blotting, and radiosensitivity testing [8]. A new rapid assay for diagnosis, which also promises to identify carriers, has recently been described [9]. No effective disease-modifying therapy is presently available, although supportive therapy includes medications that control drooling and tremors. Aggressive physical therapy prevents contractures and minimizes lung infections Speech therapy improves enunciation and, thereby, schoolwork and social relationships.

The potential benefits of glucocorticoids for A-T have been considered before. A-T patients often develop lymphoid malignancies, for which treatment regimens usually include glucocorticoids or ‘steroids’. Parents of patients receiving glucocorticoids have been quick to point out that the neurological symptoms diminish, sometimes dramatically, during treatment. This led to attempts in the 1980s to focus on the use of these drugs for long-term treatment of the neurological problems [10]. Whilst it was clear that they did reduce symptoms, it seemed equally clear that once discontinued, the symptoms reappeared. This has also been the recent experience of Buoni et al. [1], who described a 3-year-old boy with both A-T and bronchial asthma in whom the ataxia improved whenever the asthma was being treated with betamethasone (the accompanying videos were both impressive and encouraging). Broccoletti et al. [2] followed up by administering betamethasone (0.1 mg/kg/day) for 10 days to six consecutive patients ranging in age from 5–30 years. All but one improved, with the most striking improvement being observed in speech, stance and finger-chase items. Notably, the greatest clinical improvements were seen in the two younger patients, in whom MRI changes were not yet present. One patient maintained slight improvement for a week after steroids was discontinued.

The issue then becomes one of benefit-to-risk. These patients gain weight and become ‘Cushingoid’ from the steroids just as any other patient would. They are already immunocompromised by the underlying absence of ATM protein. Roughly two-thirds of A-T patients manifest both humoral and cellular immune deficiencies, including lymphopenia, reduced T and B cell responses, IgA and IgG2 deficiency, and poor antibody responses to lipopolysaccharide antigens, such as pneumococcal vaccines [1116]. Whilst the immunodeficiency has often been suggested as the cause of the increased cancer susceptibility, recent data suggest that the cancers most probably result from inefficient recognition and repair of double strand DNA breaks, leading to larger chromosomal aberrations and the development of malignant clones [1722].

Glucocorticoids are known to reduce both inflammatory and immune responses, which can increase the frequency of infections. Furthermore, bones weaken and fracture, especially in inactive patients, and growth is retarded. Thus, long-term steroid use in such patients would initially appear to be a step in the wrong direction, especially when one considers that all but the most severely affected A-T patients continue to function fairly well at home and in school until about age 8. Also, at age 8, it is still extremely difficult to predict the long-term outcome of the natural disease.

This is not to say that anti-inflammatory agents might not have a role in slowing the progressive neurodegeneration. ATM kinase, the missing protein in A-T, is thought to phosphorylate over 700 downstream substrates [23] and these proteins impact on many pathways and provide many potential targets upon which steroids might act. Apoptosis is deficient; p53 is not activated efficiently by ATM nor protected from degradation [18,22,2427]. NFκB, another transcription factor with myriad targets, is also phosphorylated by ATM kinase [28]. Both the Fois [1] and Pignata [2] groups have considered substituting other glucocorticoids that might be less toxic or perhaps cross the blood–brain barrier more efficiently. Methylprednisolone had no beneficial effect. Perhaps some of the newer anti-inflammatory agents in development may prove more suitable for long-term use in A-T patients [29].

Alternatively, could the effects of betamethasone result from its activity as an antioxidant? A-T cells react poorly to oxidative stress [30]. As well, oxidative stress plays a major role in neural degeneration [31,32]. The possible antioxidant mechanism is addressed by Russo et al. [3] in this issue. In a very limited study, the authors measured intracellular glutathione levels, reactive oxygen species (ROS) production, and lipid peroxidation in the same six A-T patients receiving betamethasone for 10 days [2]. They compared some of these results to clinical improvement, as assessed by SARA – a clinical Scale for the Assessment and Rating of Ataxia. A marked reduction in ROS levels was observed in one patient, suggesting that antioxidative mechanisms may play a role in improving the cerebellar function. However, once again, in their final figure, we see that ROS levels had returned to normal only 7 days after discontinuing the betamethasone.

The study further suggests that ROS levels might serve as a biomarker for predicting which patients might be the best candidates for betamethasone treatment. Other biomarkers to consider might be: (i) the levels of ATM kinase activity in peripheral blood lymphocytes of patients, (ii) using a kinetic response curve for the phosphorylation of SMC1 after cells have been damaged by bleomycin or irradiation [9], or 3) certain ATM mutations may render some A-T patients more responsive to steroid treatment. It is interesting that despite clinical improvement with betamethasone, the elevated serum alphafetoprotein did not change in the single patient in whom such measurements were reported [1].

As with other clinical trials for chronic diseases, it will be very difficult to select whether to use subjects who are older and more severely affected, or younger less affected children. What is most interesting about the present studies is that symptomatic neurological improvement was observed within 1 week of starting betamethasone, despite 3–21 years of previous neurological deterioration! This rapid turnabout argues strongly against the need for cellular replacements and the reestablishment of neural synapses and circuitry, which usually take months or years. The rapid reversibility also provides strong encouragement for the development of other therapeutic approaches, such as antioxidants and mutation-targeted therapy [3336]. On the other hand, the observation that older patients did not respond as well as younger ones suggests that a threshold level of Purkinje cell numbers or other cerebellar hard-wiring may be a prerequisite for successful steroid therapy in A-T [37].

Thus, on the basis of very limited studies, it would appear that steroids must remain within the armamentarium of supportive medicine for A-T until further evidence can be gathered as to their potential role as disease-modifying agents. The bailout at this juncture seems to be only a temporary holding action and the lurking costs and risks are still difficult to assess. Unraveling the molecular pathogenesis of A-T remains the most promising road to a disease-modifying treatment.

References

  • 1.Buoni S, Zanolli R, Sorrentino L, Fois A. Betamethasone and improvement of neurological symptoms in ataxia-telangiectasia. Arch Neurol. 2006;63:1479–1482. doi: 10.1001/archneur.63.10.1479. [DOI] [PubMed] [Google Scholar]
  • 2.Broccoletti T, Del Giudice E, Amorosi S, et al. Steroid-induced improvement of neurological signs in ataxia-telangiectasia patients. Eur J Neurol. 2008;15:223–228. doi: 10.1111/j.1468-1331.2008.02060.x. [DOI] [PubMed] [Google Scholar]
  • 3.Russo I, Cosentino C, Del Giudice E, et al. In ataxia-teleangiectasia betamethasone response is inversely correlated to cerebellar atrophy and directly to antioxidative capacity. Eur J Neurol. 2009;16:755–9. doi: 10.1111/j.1468-1331.2009.02600.x. [DOI] [PubMed] [Google Scholar]
  • 4.Boder E. Ataxia-telangiectasia: an overview. In: Gatti RA, Swift M, editors. Ataxia-telangiectasia: Genetics, Neuropathology, and Immunology of a Degenerative Disease of Childhood. Kroc Conference Series. Vol. 19. New York: Alan R. Liss Inc; 1985. pp. 1–63. [Google Scholar]
  • 5.Gatti RA. Ataxia-telangiectasia. In: Scriver CR, Beaudet A, Sly WS, Valle D, editors. The Metabolic and Molecular Basis of Inherited Disease. 8. New York: McGraw-Hill, Inc; 2001. pp. 705–732. [Google Scholar]
  • 6.Perlman S, Becker-Catania S, Gatti RA. Ataxia-telangiectasia: diagnosis and treatment. Semin Pediatr Neurol. 2003;10:173–182. doi: 10.1016/s1071-9091(03)00026-3. [DOI] [PubMed] [Google Scholar]
  • 7.Chun HH, Gatti RA. Ataxia-telangiectasia, an evolving phenotype. DNA Repair. 2004;3:1187–1196. doi: 10.1016/j.dnarep.2004.04.010. [DOI] [PubMed] [Google Scholar]
  • 8.Mitui M, Nahas SA, Chun HH, Gatti RA. Diagnosis of ataxia-telangiectasia: ATM mutations associated with cancer. In: Nakamura R, Grody W, Wu S, Nagle D, editors. Cancer Diagnostics: Current and Future Trends. Humana Press; Totowa, NJ: 2004. pp. 473–487. [Google Scholar]
  • 9.Nahas SA, Butch AW, Gatti RA. Rapid flow cytometry-based SMC1 phosphorylation assay for identification of ataxia-telangiectasia homozygotes and heterozygotes. Clin Chem. 2009;55:463–472. doi: 10.1373/clinchem.2008.107128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gatti RA. Concluding discussion. In: Gatti RA, Swift M, editors. Ataxia-telangiectasia: Genetics, Neuropathology, and Immunology of a Degenerative Disease of Childhood. Kroc Conference Series. Vol. 19. New York: Alan R. Liss, Inc.; 1985. pp. 357–371. [Google Scholar]
  • 11.Oxelius VA, Berkel AI, Hanson LA. IgG2 deficiency in ataxia-telangiectasia. N Engl J Med. 1982;306:515–520. doi: 10.1056/NEJM198203043060905. [DOI] [PubMed] [Google Scholar]
  • 12.Gatti RA, Bick M, Tam CF, et al. Ataxia-telangiectasia: a multiparameter analysis of eight families. Clin Immunol Immunopathol. 1982;23:501–516. doi: 10.1016/0090-1229(82)90134-9. [DOI] [PubMed] [Google Scholar]
  • 13.Woods CG, Taylor AMR. Ataxia telangiectasia in the British Isles: the clinical and laboratory features of 70 affected individuals. Q J Med New Series. 1992;298:169–179. [PubMed] [Google Scholar]
  • 14.Regueiro JR, Porras O, Lavin M, Gatti RA. Ataxia-telangiectasia. A pimary immunodeficiency revisited. Immunol Allergy Clin North Am. 2000;20:177–206. [Google Scholar]
  • 15.Sanal O, Ozbas-Gerceker F, Yel L, et al. Defective anti-polysaccharide antibody response in patients with ataxia-telangiectasia. Turk J Pediatr. 2004;46:208–213. [PubMed] [Google Scholar]
  • 16.Pashankar F, Singhal V, Akabogu I, Gatti RA, Goldman FD. Intact T cell responses in ataxia telangiectasia. Clin Immunol. 2006;120:158–162. doi: 10.1016/j.clim.2006.04.568. [DOI] [PubMed] [Google Scholar]
  • 17.Lou Z, Minter-Dykhouse K, Franco S, et al. MDC1 maintains genomic stability by participating in the amplification of ATM-dependent DNA damage signals. Mol Cell. 2006;21:187–200. doi: 10.1016/j.molcel.2005.11.025. [DOI] [PubMed] [Google Scholar]
  • 18.Morales JC, Franco S, Murphy MM, et al. 53BP1 and p53 synergize to suppress genomic instability and lymphomagenesis. Proc Natl Acad Sci USA. 2006;103:3310–3315. doi: 10.1073/pnas.0511259103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Franco S, Alt FW, Manis JP. Pathways that suppress programmed DNA breaks from progressing to chromosomal breaks and translocations. DNA Repair. 2006;5:1030–1041. doi: 10.1016/j.dnarep.2006.05.024. [DOI] [PubMed] [Google Scholar]
  • 20.Zha S, Sekiguchi J, Brush JW, Bassing CH, Alt FW. Complementary functions of ATM and H2AX in development and suppression of genomic instability. Proc Natl Acad Sci USA. 2008;105:9302–9306. doi: 10.1073/pnas.0803520105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bassing cH, Ranganath S, Murphy M, Savic V, Gleason M, Alt FW. Aberrant V(D)J recombination is not required for rapid development of H2AX/p53-deficient thymic lymphomas with clonal translocations. Blood. 2008;111:2163–2169. doi: 10.1182/blood-2007-08-104760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Goodarzi AA, Noon AT, Deckbar D, et al. ATM signaling facilitates repair of DNA double-strand breaks associated with heterochromatin. Mol Cell. 2008;31:167–177. doi: 10.1016/j.molcel.2008.05.017. [DOI] [PubMed] [Google Scholar]
  • 23.Mitzuoka S, Ballif BA, Smogorzewska A, et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science. 2007;316:1160–1166. doi: 10.1126/science.1140321. [DOI] [PubMed] [Google Scholar]
  • 24.Lee Y, Chong MJ, McKinnon PJ. Ataxia telangiectasia mutated-dependent apoptosis after genotoxic stress in the developing nervous system is determined by cellular differentiation status. J Neurosci. 2001;21:6687–6693. doi: 10.1523/JNEUROSCI.21-17-06687.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Maclean KH, Kastan MB, Cleveland JL. ATM deficiency affects both apoptosis and proliferation to augment myc-induced lymphomagenesis. Mol Cancer Res. 2007;5:705–711. doi: 10.1158/1541-7786.MCR-07-0058. [DOI] [PubMed] [Google Scholar]
  • 26.Walkley CR, Qudsi R, Sankaran VG, et al. Conditional mouse osteosarcoma, dependent on p53 loss and potentiated by loss of Rb, mimics the human disease. Genes Dev. 2008;22:1662–1676. doi: 10.1101/gad.1656808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gaul L, Mandl-Weber S, Baumann P, Emmerich B, Schmidmaier R. Bendamustine induces G2 cell cycle arrent and apoptosis in myeloma cells: the role of ATM-Chk2-Cdc25A and ATM-p53-p21-pathways. J Cancer Res Clin Oncol. 2008;134:245–253. doi: 10.1007/s00432-007-0278-x. [DOI] [PubMed] [Google Scholar]
  • 28.Yoshida K, Ozaki T, Furuya K, et al. ATM-dependent nuclear accumulation of IKK-alpha plays an important role in the regulation of p73-mediated apoptosis in response to cisplatin. Oncogene. 2008;27:1183–1188. doi: 10.1038/sj.onc.1210722. [DOI] [PubMed] [Google Scholar]
  • 29.Stahn C, Lowenberg M, Hommes DW, Buttgereit F. Molecular mechanisms of glucocorticoid action and selective glucocorticoid receptor agonists. Mol Cell Endocrinol. 2007;275:71–78. doi: 10.1016/j.mce.2007.05.019. [DOI] [PubMed] [Google Scholar]
  • 30.Barzilai A, Rotman G, Shiloh Y. ATM deficiency and oxidative stress: a new dimension of defective response to DNA damage. DNA Repair. 2002;1:3–25. doi: 10.1016/s1568-7864(01)00007-6. [DOI] [PubMed] [Google Scholar]
  • 31.Ceccatelli S, Tamm C, Zhang Q, Chen M. Mechanisms and modulation of neural cell damage induced by oxidative stress. Physiol Behav. 2007;92:87–92. doi: 10.1016/j.physbeh.2007.05.048. [DOI] [PubMed] [Google Scholar]
  • 32.Schulz JB, Lindenau J, Seyfried J, Dichganx J. Glutathi-one, oxidative stress and neurodegeneration. Eur J Biochem. 2000;267:4904–4911. doi: 10.1046/j.1432-1327.2000.01595.x. [DOI] [PubMed] [Google Scholar]
  • 33.Lavin MF, Gueven N, Bottle S, Gatti RA. Current and potential therapeutic strategies for the treatment of ataxia-telangiectasia. Br Med Bull. 2007;10:1–19. doi: 10.1093/bmb/ldm012. [DOI] [PubMed] [Google Scholar]
  • 34.Lai CH, Chun HH, Nahas SA, et al. Correction of ATM gene function by aminoglycoside-induced readthrough of premature termination codons. Proc Natl Acad Sci USA. 2004;101:15676–15681. doi: 10.1073/pnas.0405155101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Du L, Pollard J, Gatti RA. Correction of prototypic ATM splicing mutation and aberrant ATM function with antisense morpholino oligonucleotides. Proc Natl Acad Sci USA. 2007;104:6007–6012. doi: 10.1073/pnas.0608616104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hu H, Gatti RA. New approaches to treatment of primary immune deficiencies: fixing mutations with chemicals. Curr Opin Allergy Clin Immunol. 2008;8:540–546. doi: 10.1097/ACI.0b013e328314b63b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gatti RA, Vinters HV. Cerebellar pathology in ataxia-telangiectasia: the significance of basket cells. In: Gatti RA, Swift M, editors. Ataxia-telangiectasia: Genetics, Neuropathology, and Immunology of a Degenerative Disease of Childhood. Vol. 19. New York: Alan R Liss Inc; 1985. pp. 225–232. [PubMed] [Google Scholar]

RESOURCES