Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-8zxtt Total loading time: 0 Render date: 2024-07-09T10:22:22.176Z Has data issue: false hasContentIssue false

Chapter 6 - Subcutaneous, intranasal and transdermal dopamine agonists in the management of Parkinson's disease

from Section I - The Pharmacological Basis for Parkinson's Disease Treatment

Published online by Cambridge University Press:  05 March 2016

Néstor Gálvez-Jiménez
Affiliation:
Cleveland Clinic, Florida
Hubert H. Fernandez
Affiliation:
Cleveland Clinic, Ohio
Alberto J. Espay
Affiliation:
University of Cincinnati
Susan H. Fox
Affiliation:
Toronto Western Hospital
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Parkinson's Disease
Current and Future Therapeutics and Clinical Trials
, pp. 48 - 62
Publisher: Cambridge University Press
Print publication year: 2016

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Kvernmo, T, Härtter, S, Burger, E. A review of the receptor-binding and pharmacokinetic properties of dopamine agonists. Clin Ther 2006; 28: 106578.CrossRefGoogle ScholarPubMed
Jenner, P. Preventing and controlling dyskinesia in Parkinson's disease – a view of current knowledge and future opportunities. Mov Disord 2008; 23 (Suppl. 3): S5858.Google Scholar
Chase, TN, Baronti, F, Fabbrini, G, et al. Rationale for continuous dopaminomimetic therapy of Parkinson's disease. Neurology 1989; 39 (Suppl. 2): 710.Google ScholarPubMed
Mouradian, MM, Heuser, IJ, Baronti, F, Chase, TN. Modification of central dopaminergic mechanisms by continuous levodopa therapy for advanced Parkinson's disease. Ann Neurol 1990; 27: 1823.Google Scholar
Brotchie, J, Jenner, P. New approaches to therapy. In: Brotchie, J, Bezard, E, Jenner, P, eds. Pathophysiology, Pharmacology, and Biochemistry of Dyskinesia. San Diego: Elsevier Academic Press, 2011; 12350.CrossRefGoogle Scholar
Chaudhuri, KR, Schapira, AH. Non-motor symptoms of Parkinson's disease: dopaminergic pathophysiology and treatment. Lancet Neurol 2009; 8: 46474.CrossRefGoogle ScholarPubMed
Storch, A, Schneider, CB, Wolz, M, et al. Nonmotor fluctuations in Parkinson disease: severity and correlation with motor complications. Neurology 2013; 80 : 8009.CrossRefGoogle ScholarPubMed
Ray Chaudhuri, K, Rizos, A, Sethi, K. Motor and nonmotor complications in Parkinson's disease: an argument for continuous drug delivery? J Neural Transm 2013; 120: 130520.CrossRefGoogle Scholar
Mouradian, MM. Scientific rationale for continuous dopaminergic stimulation in Parkinson's disease. Eur Neurol Rev 2006; 626.Google Scholar
Marinnan, S, Emmanuel, A, Burn, D. Delayed gastric emptying in Parkinson's disease. Mov Disord 2014; 29: 2332.Google Scholar
Naidu, Y, Ray Chaudhuri, K. Transdermal rotigotine: a new non-ergot dopamine agonist for the treatment of Parkinson's disease. Expert Opin Drug Deliv 2007; 4: 11118.CrossRefGoogle ScholarPubMed
Jenner, P. A novel dopamine agonist for the transdermal treatment of Parkinson's disease. Neurology 2005; 65 (Suppl. 1): S35.CrossRefGoogle ScholarPubMed
Paul, ML, Graybiel, AM, David, JC, Robertson, HA. D1-like and D2-like dopamine receptors synergistically activate rotation and c-fos expression in the dopamine-depleted striatum in a rat model of Parkinson's disease. J Neurosci 1992; 12: 372942.Google Scholar
Albanese, A, Jenner, P, Marsden, CD, Stephenson, JD. Bladder hyperreflexia induced in marmosets by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Neurosci Lett. 1988; 87: 4650.Google Scholar
Tomiyama, M, Kimura, T, Maeda, T, et al. A serotonin 5-HT1A receptor agonist prevents behavioral sensitization to L-DOPA in a rodent model of PD. Neurosci Res 2005; 52: 18594.CrossRefGoogle Scholar
Srinivasan, J, Schmidt, WJ. Treatment with α2 – adrenoceptor antagonists, 2 methoxy idazaoxan, protects 6- hydroxydopamine induced Parkinsonian symptoms in rats: neurochemical and behavioral evidence. Behav Brain Res 2004; 154: 35363.CrossRefGoogle ScholarPubMed
Cawello, W, Braun, M, Boekens, H. Absorption, disposition, metabolic fate, and elimination of the dopamine agonist rotigotine in man: administration by intravenous infusion or transdermal delivery drug metabolism and disposition. Drug Metab Dispos 2009; 37: 205560.Google Scholar
LeWitt, PA, Lyons, KE, Pahwa, R, SP 650 Study Group. Advanced Parkinson disease treated with rotigotine transdermal system: PREFER study. Neurology 2007; 68: 12627.Google Scholar
Poewe, WH, Rascol, O, Quinn, N, et al. Efficacy of pramipexole and transdermal rotigotine in advanced Parkinson's disease: a double-blind, double-dummy, randomised controlled trial. Lancet Neurol 2007; 6: 51320.Google Scholar
Trenkwalder, C, Kies, B, Rudzinska, M, et al. Rotigotine effects on early morning motor function and sleep in Parkinson's disease: a double-blind, randomized, placebo-controlled study (RECOVER). Mov Disord 2011; 26: 909.CrossRefGoogle ScholarPubMed
Ray Chaudhuri, K, Martinez-Martin, P, Antonini, A, et al. Rotigotine and specific non-motor symptoms of Parkinson's disease: post hoc analysis of RECOVER. Parkinsonism Relat Disord 2013; 19: 6605.Google Scholar
Kassubek, J, Ray Chaudhuri, K, Zesiewicz, T, et al. Rotigotine transdermal system and evaluation of pain in patients with Parkinson's disease: a post hoc analysis of the RECOVER study. BMC Neurology 2014; 14: 42.Google Scholar
Trenkwalder, C, Kies, B, Dioszeghy, P, et al. Rotigotine transdermal system for the management of motor function and sleep disturbances in Parkinson's disease: results from a 1-year, open-label extension of the RECOVER study. Basal Ganglia 2012; 2: 7985.Google Scholar
Chen, JJ, Swope, DM, Dashtipour, K, Lyons, KE. Transdermal rotigotine: a clinically innovative dopamine-receptor agonist for the management of Parkinson's disease. Pharmacotherapy 2009; 29: 145267.CrossRefGoogle ScholarPubMed
Metta, V, Muzerengi, S, Ray Chadhuri, K. Rotigotine: first dopamine agonist transdermal patch. Prescriber 2007; 18: 1930.Google Scholar
Weintraub, D, Nirenberg, MJ. Impulse control and related disorders in Parkinson's disease. Neurodegener Dis 2013; 11: 6371.Google Scholar
Rizos, A, Martinez-Martin, P, Martin, A, et al. European multicentre survey of tolerability rates and impulse control behaviour trends of prolonged release dopamine agonists in young and old PD. Mov Disord 2012; 27 (Suppl. 1): 161.Google Scholar
Priano, L, Albani, G, Brioschi, A, et al. Transdermal Apomorphine permeation from microemulsions: a new treatment in Parkinson's disease. Mov Disord 2004; 19: 93742.CrossRefGoogle ScholarPubMed
Woitalla, D, Müller, T, Benz, S, Horowski, R, Przuntek, H. Transdermal lisuride delivery in the treatment of Parkinson's disease. J Neural Transm Suppl 2004; 68: 8995.CrossRefGoogle Scholar
Dimitrova, T, Bara-Jiminez, W, Thomas, M, et al. Continuous dopaminergic stimulation with lisuride TTS (patch) in moderately advanced parkinsonian patients. Neurology 2006; 66 (Suppl. 2): A185.Google Scholar
Corrodi, H, Farnebo, LO, Fuxe, K, Hamberger, B, Ungerstedt, U. ET 495 (piribedil) and brain catecholamine mechanisms: evidence for stimulation of dopamine receptors. Eur J Pharmacol 1972; 20: 195204.Google Scholar
Montastruc, JL, Ziegler, M, Rascol, O, Malbezin, M. A randomized, double-blind study of a skin patch of a dopaminergic agonist, piribedil, in Parkinson's disease. Mov Disord 1999; 14: 33641.Google Scholar
Kapoor, R, Turjanski, N, Frankel, J, et al. Intranasal apomorphine: a new treatment in Parkinson's disease. J Neurol Neurosurg Psychiatry 1990; 53: 1015.CrossRefGoogle ScholarPubMed
Kleedorfer, B, Turjanski, N, Ryan, R, et al. Intranasal apomorphine in Parkinson's disease. Neurology 1991; 41: 7612.Google Scholar
van Laar, T, Jansen, EN, Essink, AW, Neef, C. Intranasal apomorphine in parkinsonian on–off fluctuations. Arch Neurol 1992; 49: 4824.Google Scholar
Sam, E, Jeanjean, AP, Maloteaux, JM, Verbeke, N. Apomorphine pharmacokinetics in parkinsonism after intranasal and subcutaneous application. Eur J Drug Metab Pharmacokinet. 1995; 20: 2733.Google Scholar
Dewey, RB Jr, Maraganore, DM, Ahlskog, JE, Matsumoto, JY. Intranasal apomorphine rescue therapy for parkinsonian “off” periods. Clin Neuropharmacol 1996; 19: 193201.Google Scholar
Esteban Muñoz, J, Martí, MJ, Marín, C, Tolosa, E. Long-term treatment with intermitent intranasal or subcutaneous apormorphine in patients with levodopa-related motor fluctuations. Clin Neuropharmacol 1997; 20: 24552.Google Scholar
Dewey, RB Jr, Maraganore, DM, Ahlskog, JE, Matsumoto, JY. A double-blind, placebo-controlled study of intranasal apomorphine spray as a rescue agent for off-states in Parkinson's disease. Mov Disord 1998; 13: 7827.Google Scholar
Wickremaratchi, M, Hadjikoutis, S, Weiser, R, et al. Efficacy and tolerability of intranasal apomorphine powder (INAP) 5mg as rescue therapy in subjects with Parkinson's disease (PD) complicated by motor fluctuations. J Neurol Neurosurg Psychiatry 2003; 74: 144860.Google Scholar
Deleu, D, Hanssens, Y, Northway, MG. Subcutaneous apomorphine: an evidence-based review of its use in Parkinson's disease. Drugs Aging 2004; 21: 687709.CrossRefGoogle ScholarPubMed
Schwab, RS, Amador, LV, Lettvin, JY. Apomorphine in Parkinson's disease. Trans Am Neurol Assoc 1951; 56: 2513.Google ScholarPubMed
Cotzias, GC, Papavasiliou, PS, Tolosa, ES, Mendez, JS, Bell-Midura, M. Treatment of Parkinson's disease with apomorphines. Possible role of growth hormone. The N Engl J Med 1976; 294: 56772.Google Scholar
Stibe, CM, Lees, AJ, Kempster, PA, Stern, GM. Subcutaneous apomorphine in parkinsonian on–off oscillations. Lancet 1988; 331: 4036.Google Scholar
Chaudhuri, KR, Critchley, P, Abbott, RJ, Pye, IF, Millac, PAH. Subcutaneous apomorphine for on–off oscillations in Parkinson's disease. Lancet 1988; 332: 1260.Google Scholar
LeWitt, PA. Subcutaneously administered apomorphine: pharmacokinetics and metabolism. Neurology 2004; 62 (Suppl. 4): S811.Google Scholar
Nicolle, E, Pollak, P, Serre-Debeauvais, F, et al. Pharmacokinetics of apomorphine in parkinsonian patients. Fundam Clin Pharmacol 1993; 7: 24552.Google Scholar
Manson, AJ, Hanagasi, H, Turner, K, et al. Intravenous apomorphine therapy in Parkinson's disease: clinical and pharmacokinetic observations. Brain 2001; 124: 33140.Google Scholar
Hofstee, DJ, Neef, C, van Laar, T, Jansen, EN. Pharmacokinetics of apomorphine in Parkinson's disease: plasma and cerebrospinal fluid levels in relation to motor responses. Clin Neuropharmacol 1994; 17: 4552.Google Scholar
Frankel, JP, Lees, AJ, Kempster, PA, Stern, GM. Subcutaneous apomorphine in the treatment of Parkinson's disease. J Neurol Neurosurg Psychiatry 1990; 53: 96101.Google Scholar
Pollak, P, Champay, AS, Gaio, JM, et al. [Subcutaneous administration of apomorphine in motor fluctuations in Parkinson's disease]. Rev Neurol (Paris) 1990; 146 : 11622 (in French).Google Scholar
Hughes, AJ, Bishop, S, Kleedorfer, B, et al. Subcutaneous apomorphine in Parkinson's disease: response to chronic administration for up to five years. Mov Disord 1993; 8: 16570.Google Scholar
Stocchi, F, Bramante, L, Monge, A, et al. Apomorphine and lisuride infusion. A comparative long-term study. Adv Neurol 1993; 60: 6535.Google Scholar
Poewe, W, Kleedorfer, B, Wagner, M, Bosch, S, Schelosky, L. Continuous subcutaneous apomorphine infusions for fluctuating Parkinson's disease. Long-term follow-up in 18 patients. Adv Neurol 1993; 60: 6569.Google ScholarPubMed
Kreczy-Kleedorfer, B, Wagner, M, Bösch, S, Poewe, W. [Long-term results of continuous subcutaneous apomorphine pump therapy in patients with advanced Parkinson disease]. Nervenarzt 1993; 64: 2215 (in German).Google Scholar
Gancher, ST, Nutt, JG, Woodward, WR. Apomorphine infusional therapy in Parkinson's disease: clinical utility and lack of tolerance. Mov Disord 1995; 10: 3743.Google Scholar
Colzi, A, Turner, K, Lees, AJ. Continuous subcutaneous waking day apomorphine in the long term treatment of levodopa induced interdose dyskinesias in Parkinson's disease. J Neurol Neurosurg Psychiatry 1998; 64: 5736.Google Scholar
Pietz, K, Hagell, P, Odin, P. Subcutaneous apomorphine in late stage Parkinson's disease: a long-term follow-up. J Neurol Neurosurg Psychiatry 1998; 65: 70916.Google Scholar
Wenning, GK, Bösch, S, Luginger, E, Wagner, M, Poewe, W. Effects of long-term, continuous subcutaneous apomorphine infusions on motor complications in advanced Parkinson's disease. Adv Neurol 1999; 80: 5458.Google Scholar
Stocchi, F, Vacca, L, De Pandis, MF, et al. Subcutaneous continuous apomorphine infusion in fluctuating patients with Parkinson's disease: long-term results. Neurol Sci. 2001; 22: 934.CrossRefGoogle ScholarPubMed
Kanovsky, P, Kubova, D, Bares, M, et al. Levodopa-induced dyskinesias and continuous subcutaneous infusions of apomorphine: results of a two-year, prospective follow-up. Mov Disord 2002; 17: 18891.Google Scholar
Manson, AJ, Turner, K, Lees, AJ. Apomorphine monotherapy in the treatment of refractory motor complications of Parkinson's disease: long-term follow-up study of 64 patients. Mov Disord 2002; 17: 123541.Google Scholar
Di Rosa, AE, Epifanio, A, Antonini, A, et al. Continuous apomorphine infusion and neuropsychiatric disorders: a controlled study in patients with advanced Parkinson's disease. Neurol Sci 2003; 24: 1745.CrossRefGoogle ScholarPubMed
Morgante, L, Basile, G, Epifanio, A, et al. Continuous apomorphine infusion (CAI) and neuropsychiatric disorders in patients with advanced Parkinson's disease: a follow-up of two years. Arch Gerontol Geriatr Suppl 2004; 9: 2916.Google Scholar
Tyne, HL, Parsons, J, Sinnott, A, et al. A 10 year retrospective audit of long-term apomorphine use in Parkinson's disease. J Neurol 2004; 251: 13704.CrossRefGoogle ScholarPubMed
Katzenschlager, R, Hughes, A, Evans, A, et al. Continuous subcutaneous apomorphine therapy improves dyskinesias in Parkinson's disease: a prospective study using single-dose challenges. Mov Disord 2005; 20: 1517.Google Scholar
De Gaspari, D, Siri, C, Landi, A, et al. Clinical and neuropsychological follow up at 12 months in patients with complicated Parkinson's disease treated with subcutaneous apomorphine infusion or deep brain stimulation of the subthalamic nucleus. J Neurol Neurosurg Psychiatry 2006; 77: 4503.Google Scholar
Garcia Ruiz, PJ, Sesar Ignacio, A, Ares Pensado, B, et al. Efficacy of long-term continuous subcutaneous apomorphine infusion in advanced Parkinson's disease with motor fluctuations: a multicenter study. Mov Disord 2008; 23: 11306.CrossRefGoogle ScholarPubMed
Martinez-Martin, P, Reddy, P, Antonini, A, et al. Chronic subcutaneous infusion therapy with apomorphine in advanced Parkinson's disease compared to conventional therapy: a real-life study of non-motor effect. J Parkinson's Dis 2011; 1: 197203.Google Scholar
Antonini, A, Isaias, IU, Rodolfi, G, et al., A 5-year prospective assessment of advanced Parkinson disease patients treated with subcutaneous apomorphine infusion or deep brain stimulation, J Neurol 2011; 258: 57985.Google Scholar
Drapier, S, Gillioz, AS, Leray, E, et al. Apomorphine infusion in advanced Parkinson's patients with subthalamic stimulation contraindications. Parkinsonism Relat Disord 2012; 18: 404.Google Scholar
Alegret, M, Valldeoriola, F, Marti, M, et al. Comparative cognitive effects of bilateral subthalamic stimulation and subcutaneous continuous infusion of apomorphine in Parkinson's disease. Mov Disord 2004; 19: 14639.CrossRefGoogle ScholarPubMed
Todorova, A Ray Chaudhuri, K. Subcutaneous apomorphine and non motor symptoms in Parkinson's disease. Parkinsonism Relat Disord 2013; 19: 10738.Google Scholar
Ray Chaudhuri, K, Abbott, RJ, Millac, PAH. Subcutaneous apomorphine for parkinsonian patients with psychiatric side effects on oral treatment. J Neurol Neurosurg Psychiatry 1991; 54: 3723.Google Scholar
Ellis, C, Lemmens, G, Parkes, JD, et al. Use of apomorphine in parkinsonian patients with neurospsychiatric complications of oral treatment. Parkinsonism Relat Disord 1997; 3: 1037.Google Scholar
van Laar, T, Postuma, AG, Drent, M. Continuous subcutaneous infusion of apomorphine can be used safely in patients with Parkinson's disease and pre-existing visual hallucinations. Parkinsonism Relat Disord 2010; 16: 712.Google Scholar
Issacson, S, Ray Chaudhuri, K. Morning akinesia and the potential role of gastroparesis – managing delayed onset of first daily dose of oral levodopa in patients with Parkinson's disease. European Neurological Review 2013; 8: 824.CrossRefGoogle Scholar
Mathers, SE, Kempster, PA, Law, PJ, et al. Anal sphincter dysfunction in Parkinson's disease. Arch Neurol 1989; 46: 10614.Google Scholar
Edwards, LL, Quigley, EM, Harned, RK, Hofman, R, Pfeiffer, RF. Defecatory function n Parkinson's disease: response to apomorphine. Ann Neurol 1993; 33: 4903.Google Scholar
Tison, F, Wiart, L, Guatterie, M, et al. Effects of central dopaminergic stimulation by apomorphine on swallowing disorders in Parkinson's disease. Mov Disord 1996; 11: 72932.Google Scholar
Reuter, I, Ellis, CM, Ray Chaudhuri, K. Nocturnal subcutaneous apomorphine infusion in Parkinson's disease and restless legs syndrome. Acta Neurol Scand 1999; 100: 1637.Google Scholar
Tribl, GG, Sycha, T, Kotzailias, N, Zeitlhoger, J, Auff, E. Apomorphine in idiopathic restless legs syndrome: an exploratory study. J Neurol Neurosurg Psychiatry 2005; 76: 1815.CrossRefGoogle ScholarPubMed
Magennis, B, Cashell, A, O'Brien, D, Lynch, T. An audit of apomorphine in the management of complex idiopathic Parkinson's disease in Ireland. Mov Disord 2012; 27 (Suppl. 1): S44.Google Scholar
Todorova, A, Martin, A, Okai, D, et al. Assessment of impulse control disorders in Parkinson's patients with infusion therapies: a single centre experience. Mov Disord 2013; 28 (Suppl. 1): S133.Google Scholar
Corsini, GU, Del Zompo, M, Gessa, GL, Mangoni, A. Therapeutic efficacy of apomorphine combined with an extracerebral inhibitor of dopamine receptors in Parkinson's disease. Lancet 1979; 313: 9546.Google Scholar
Stocchi, F, Ruggieri, S, Antonini, A, et al. Subcutaneous lisuride infusion in Parkinson's disease: clinical results using different modes of administration. J Neural Transm 1988; 27: 2733.Google Scholar
Krause, W, Nieuweboer, B, Ruggieri, S, Stocchi, F, Suchy, I. Pharmacokinetics of lisuride after subcutaneous infusion. J Neural Transm Suppl 1988; 27: 714.Google Scholar
Stocchi, F, Ruggieri, S, Vacca, L, Olanow, CW. Prospective randomized trial of lisuride infusion versus oral levodopa in patients with Parkinson's disease. Brain 2002; 125: 205866.Google Scholar
Heinz, A, Suchy, I, Klewin, I, et al. Long-term observation of chronic subcutaneous administration of lisuride in the treatment of motor fluctuations in Parkinson's disease. J Neural Transm Park Dis Dement Sect 1992; 4: 291301.Google Scholar
Obeso, JA, Luquin, MR, Vaamonde, J, Martînez Lage, JM. Subcutaneous administration of lisuride in the treatment of complex motor fluctuations in Parkinson's disease. J Neural Transm Suppl 1988; 27: 1725.Google Scholar
Critchley, PH, Grandas Perez, F, Quinn, NP, Parkes, JD, Marsden, CD. Continuous subcutaneous lisuride infusions in Parkinson's disease. J Neural Transm Suppl. 1988; 27: 5560.Google Scholar
Vaamonde, J, Luquin, MR, Obeso, JA. Subcutaneous lisuride infusion in Parkinson's disease. Response to chronic administration in 34 patients. Brain 1991; 114: 60117.Google Scholar
Hayashi, R, Tako, K, Makishita, H, Koyama, J, Yanagisawa, N. Efficacy of a low-dose subcutaneous lisuride infusion in Parkinson's disease. Intern Med 1998; 37: 4448.Google Scholar
Hofmann, C, Penner, U, Dorow, R, et al. Lisuride, a dopamine receptor agonist with 5-HT2B receptor antagonist properties: absence of cardiac valvulopathy adverse drug reaction reports supports the concept of a crucial role for 5-HT2B receptor agonism in cardiac valvular fibrosis. Clin Neuropharmacol 2006; 29: 806.Google Scholar
Canesi, M, Mariani, C, Isaias, IU, Pezzoli, G. Night-time use of Rotigotine in advanced Parkinson's disease. Funct Neurol 2010; 25: 2013.Google Scholar
Todorova, A, Martinez-Martin, P, Martin, A, et al. Daytime apomorphine infusion combined with transdermal Rotigotine patch therapy is tolerated at 2 years: a 24-h treatment option in Parkinson's disease. Basal Ganglia 2013; 3: 12730.CrossRefGoogle Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×