Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2019 Jul 22;85(10):2213–2217. doi: 10.1111/bcp.14032

Why maximum tolerated dose?

Hans G Stampfer 1,, Genevieve M Gabb 2, Simon B Dimmitt 3,4
PMCID: PMC6783596  PMID: 31219196

Abstract

A long‐established approach to the pharmacological treatment of disease has been to start low and go slow. However, clinicians often prescribe up to maximum tolerated dose (MTD), especially when treating acute and more severe disease, without evidence to show that MTD is more likely to improve outcomes. Cardiovascular guidelines for some indications advocate MTD even in prevention, for example hypercholesterolaemia, without compelling evidence of better outcomes. This review explores the origins and potential problems of prescribing medications at MTD. Oral effective dose 50 (ED50) may be a useful guide for balancing efficacy and safety.

Keywords: clinical pharmacology, dosing outcomes, effective dose 50, maximum tolerated dose

1. INTRODUCTION

The common practice of prescribing the maximum tolerated dose (MTD) of drugs such as antidepressants, antipsychotics, stimulants, statins,1, 2 vasodilators and β‐blockers3 is not evidence based. The origins of MTD can be traced to animal toxicity testing in drug development4 after a pharmacologically active dose has been established.5 Different definitions of MTD have emerged4 since Sontag's original proposal: “the highest dose of the test agent given during the chronic study that can be predicted not to alter the animals’ normal longevity from effects other than carcinogenicity.”6 Early phase human clinical trials attempted to establish the highest dose of a drug that does not cause overt toxicity or unacceptable adverse effects within a specified time period.

Clinicians often base dose on disease severity and MTD may be presumed necessary or worth trialling in serious or worsening conditions such as malignant and cardiovascular disease, vasculitis, epilepsy and serious mental illness. However, there is no evidence for better clinical outcomes with MTD and harms may overtake benefit as dose is increased. Continuation of a dose increase is justified only if there is unequivocal evidence of improved efficacy, along with acceptable adverse effects, especially in long‐term treatment of chronic disease.

Benefit may be difficult to weigh against the risk of harms with dose increases up to MTD. Effective dose 50 (ED50) defines the centre of the dose response curve, the mean population dose required to achieve half the maximum drug effect, 50% of maximum effective dose.7 ED50 may be based on measurement of biochemical or clinical endpoints and may be a useful guide to clinicians. ED50 may not be well defined in relation to clinical outcomes as, in much drug development, one dose becomes favoured in Phase I and later human trials. Investigation of the effects and outcomes on at least 3 different doses is required to estimate ED50.

With statins, coronary event reduction plateaus just above ED50 whilst a range of adverse drug reactions (ADRs) continue to increase.8 Lipid guidelines in North America have noted since 2013 that cholesterol targets have no evidence base,1 yet along with European guidelines2 promote MTD as a therapeutic target despite known reduced statin adherence related to adverse effects,9 which have been shown to be dose‐related.8 The decline in the mortality benefit on higher doses of statins8 suggests that some ADRs may contribute to mortality, for example from liver dysfunction, rhabdomyolysis, renal impairment in patients with pre‐existent renal disease10 and cerebral haemorrhage, especially in patients with previous stroke,11 which is of particular concern in the elderly.

Antihypertensive drugs are often used at higher doses than were employed in RCTs (Table 1), and aimed at epidemiological targets, which fail to factor in adverse effects of drugs. Heart failure guidelines recommend MTD of angiotensin‐converting enzyme inhibitors, angiotensin receptor blockers and β‐blockers,3 for example up to 7‐fold the ED50 of metoprolol and 16‐fold the ED50 of candesartan, despite the inevitable increase in ADRs. Cross‐sectional observational data may appear to suggest greater benefit with higher dosing but are potentially confounded by the greater tolerance of higher doses in patients with less severe disease. In contrast, no reduction in total mortality, which provides a useful summary of efficacy and safety, is evident when dose allocation (to higher or lower doses) of statins8 and angiotensin‐converting enzyme inhibitors, angiotensin receptor blockers or β‐blockers was randomised.20, 21

Table 1.

Approved, maximum and ED50 doses of common central nervous system and cardiovascular system drugs

Daily dose
Drug Maximum reported (mg) Approved Mean population
Oral ED50 dose (estimated)
(mg) (mg)
(as ratio of ED50)
Selective serotonin reuptake inhibitors 12
Citalopram 80 10–40 (3–14) 3
Sertraline 200 50–100 (5–11) 9
Fluoxetine 80 10–60 (3–20) 3
Paroxetine 60 10–40 (2–8) 5
Venlafaxine 400 37.5–225 (6–38) 6
Antipsychotics 13
Chlorpromazine 2000 25–100 (0.2–0.7) 150
Haloperidol 20 0.5–2.5 (0.5–2.5) 1
Quetiapine 800 25–200 (0.2–1.3) 150
Risperidone 16 0.5–2 (0.2–1.0) 2
Olanzapine 60 2.5–20 (0.3–2.2) 9
Stimulants
Methylphenidate 14 100 5–60 (0.5–6) 10
Dexamphetamine 15 40 5–20 (0.5–2) 10
Beta‐blockers 16
Propranolol 320 10–160 (0.2–4) 40
Metoprolol 400 50–100 (0.8–2.2) 60
Diuretics
Hydrochlorothiazide 14 100 12.5–50 (1.2–5) 10
Frusemide 12 500 20–40 (2–4) 10
Angiotensin‐converting enzyme inhibitors 17
Captopril 300 12.5–50 (0.6–2.5) 20
Perindopril 32 2–8 (0.5–2.0) 4
Statins 8
Simvastatin 160 10–80 (0.6–5) 15
Atorvastatin 80 10–80 (5–40) 2
Rosuvastatin 40 5–40 (5–40) 1
Warfarin 18 20 2–8 a (0.3–1.3) 6 b
Thyroxine 19 0.2 0.05–0.1 (0.7–1.3) 0.075 c
a

25th–75th centile dose.

b

median population dose.

c

mean population dose.

2. ADRS

Drugs usually target one benefit but may cause or contribute to a variety of adverse effects. Efficacy plateaus above ED50, whereas the incidence and severity of ADRs generally increase with dose,22 [Ferner, Aronson, this issue BJCP], exponentially with anticoagulants.23 Some ADRs are related to known drug actions, for example related to hypotension in the treatment of blood pressure or hypoglycaemia in the treatment of diabetes. However, many ADRs are less well understood. Toxicity symptoms may be delayed22 or subtle, for example, liver and renal toxicity. Clinicians should always be on high alert for ADRs, as prescribed drugs contribute to or cause 7–16% of deaths in hospitalised adults.24, 25, 26, 27

3. CLINICAL DOSING

Optimal dosing aims to maximize benefit and minimize harm and may vary between patients in acute, maintenance and preventive therapy. The temptation to increase to MTD arises in acute, serious and severe illness. Symptoms, vital signs and other surrogate indicators can provide a guide to titrating dose in both clinical trials and practice. Difficulties arise when dosing is based purely on symptoms such as pain or reported distress, commonly in psychiatry. Without objective indicators against which to titrate, doses vary widely based on a clinician's impression of dose–response. For example, whilst the recommended daily dose of desvenlafaxine is 50 mg, clinicians have prescribed up to 400 mg, many‐fold ED50 (Table 1), despite the evidence of increased ADRs28 and no reliable evidence of additional benefits. The failure to demonstrate greater clinical effectiveness with higher doses of first‐generation antipsychotics and the high rates of serious adverse effects drove their dose reduction (Table 1) and the development of second‐generation agents. However, second generation antipsychotics have also been prescribed above the recommended dose, for example up to 60 mg daily of olanzapine (Table 1), despite well documented cardiovascular29, 30 and other adverse effects.30

Clinical trials show that antidepressants are no better than placebo for mild depression and only marginally better than placebo for more serious depression27 but are often prescribed at many‐fold their ED50 (Table 1). There is growing recognition that between 20 and 30% of more serious mental illness does not respond appreciably to pharmacotherapy.31, 32 This has important implications for dosing. With only clinical indicators as a guide, clinicians may continue to increase the dose in unresponsive patients in the hope of a response that may not eventuate. There is little evidence of greater efficacy with higher doses of antidepressants33 or antipsychotics,34 whereas ADRs generally continue to increase with dose.22 Adverse effects reduce adherence.35 Patients should probably not be left on psychotropics, particularly at higher dose, without a trial of lower doses or deprescribing.36 Close consideration should be given to the use of antipsychotics in the elderly and in dementia, particularly their off‐label use for sedation and behavioural disturbance.37 Published data show that compared to placebo, harms exceed benefit38 and mortality is higher39 on antipsychotics.

An established approach to dosing, particularly for chronic diseases such as hypertension, is to start low and go slow,40 especially in the elderly. This approach facilitates dose titration to optimize effectiveness and tolerability. However, less cautious approaches may be adopted because of patient, family and community expectations of rapid results, which may drive therapeutic impatience and commencement of higher drug doses.

Safety concerns in the post‐marketing period with widely used medications have led to proactive approaches to drug safety41 such as Risk Evaluation and Mitigation Strategies42 and risk management plans (RMPs)43 for newer drugs at the time of entry to the market (Food and Drug Administration, European Medicines Agency). However, many postmarketing safety issues cannot be identified or anticipated in RMPs, with almost half of the variations in product information concerning safety aspects not envisaged by the RMP.44 These approaches may paradoxically promote a false sense of safety and lead physicians to think less critically about tempering dose.

4. DOSING AND OUTCOMES

There is a paucity of reliable data on dose‐related outcomes, even for commonly prescribed drugs. Safety and the relationship of tolerability to compliance in chronic disease has driven research in some areas to establish essential or sufficient dose, between pharmacologically active dose and no observable adverse effect level.5 Much researched examples include low‐dose aspirin45 for the prevention of stroke and coronary events and low dose methotrexate for rheumatoid arthritis,46 [Lucas, Dimmitt, Martin, this themed issue BJCP]. Metformin and paracetamol (both with ED50s around 2 g) are never recommended above their ED50. Colchicine, used in acute gout for many years with limited evidence on which to base dosing47 is a good historical example of MTD dosing. Earlier dosing regimens consisted of an immediate dose (1 mg), the usual dose threshold, with continued dosing (0.5 mg) at 1–2‐hourly intervals until the development of toxicity (diarrhoea, nausea), or symptom relief. However, the first controlled study in acute gout48 demonstrated toxicity in all patients given colchicine (mean dose 6.7 mg) with diarrhoea after a median of 24 hours, usually before the benefit of pain relief emerged. A subsequent dose comparison study demonstrated no greater benefit but only increased harms with high‐dose (4.8 mg) compared with low‐dose (1.8 mg) colchicine.49

Many patients have been prescribed up to 500 mg frusemide daily (Table 1) although safety and clinical benefits above 80 mg daily are dubious. The mean population oral ED50 is <10 mg daily.50 Higher doses do not appear to confer greater benefit even in heart failure51 and particularly with the advent of other effective coadministered antifailure medications, most patients are controlled with smaller doses of frusemide, for example 20–40 mg daily. Thiazide diuretics can cause hypokalaemia, hyponatraemia, renal impairment and increase the risk of gout and diabetes, all at least partly dose related. Doses of hydrochlorothiazide have consequently fallen from up to 100 mg daily down to 6.25–12.5 mg,52 which is around the ED50 (Table 1). Beta‐blockers confer most benefits in left ventricular failure at doses around or below ED50,21 for example 25–50 mg of metoprolol. Higher doses are less well tolerated and slower heart rate can increase systolic blood pressure, potentially negating benefits.21

The imperative to prescribe doses near or up to MTD in symptomatic and ill patients is understandable but is not supported in most instances by reliable evidence and particularly in the elderly, is unwise. More research on dose‐related outcomes is required to establish guidelines that clinicians can adopt more confidently.

5. CONCLUSION

Higher drug doses may be necessary in acute or severe illness but, when patients are stable, lower doses often suffice to ensure effective maintenance treatment, tolerability, minimisation of adverse effects and long‐term adherence. MTD as established in drug development prior to human clinical trials appears likely to come with unacceptable risks and little gain if used as a clinical dosing strategy. MTD is not justified in prevention where any adverse effects may be unacceptable. Without reliable evidence of the optimal population dose with respect to clinical outcomes and survival, clinicians may be left with limited guidance for managing patients. Mean population ED50 may be a reasonable starting dose, with due allowance for body size. The dose can be increased above ED50 if greater efficacy can be demonstrated and lowered should ADRs develop. In most illness, there is merit in maintaining patients on the lowest effective dose, with close clinical monitoring for efficacy, tolerability and adverse events.

COMPETING INTERESTS

There are no competing interests to declare.

Stampfer HG, Gabb GM, Dimmitt SB. Why maximum tolerated dose? Br J Clin Pharmacol. 2019; 85: 2213–2217. 10.1111/bcp.14032

REFERENCES

  • 1. Stone NJ, Robinson J, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular disease risk in adults: a report of the American College of Cardiology/American Heart Association task force on practice guidelines. Circulation. 2014;124:S1‐S45. [DOI] [PubMed] [Google Scholar]
  • 2. Catapano AL, Graham I, De Backer G, et al. 2016 ESC/EAS guidelines for the management of dyslipidaemias. Euro Heart J. 2016;37(39):2999‐3058. [DOI] [PubMed] [Google Scholar]
  • 3. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: the task force for the diagnosis of acute and chronic heart failure of the European Society of Cardiology (ESC) developed with the special contribution of the heart failure association (HFA) of the ESC. Eur Heart J. 2016;37:2129‐2200. [DOI] [PubMed] [Google Scholar]
  • 4. Buckley LA, Dorato MA. High dose selection in general toxicity for drug development: a pharmaceutical industry perspective. Regul Toxicol Pharmacol. 2009;54(3):301‐307. [DOI] [PubMed] [Google Scholar]
  • 5. van Gerven J, Cohen A. Integrating data from the investigational medicinal product dossier/investigator brochure. A new tool for translational integration of preclinical effects. Br J Clin Pharmacol. 2018;84(7):1457‐1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Sontag JM, Page NP, Saffiott U. Guidelines for carcinogen bioassay in small rodents. Technical Report Series. Natinoal Cancer Institute Carcinogenesis. 1976; No 1. February. [PubMed]
  • 7. Dimmitt S, Stampfer H, Martin JH. When less is more – efficacy with less toxicity at the ED50. Br J Clin Pharmacol. 2017;83(7):1365‐1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Dimmitt SB, Stampfer HG, Warren JB. The pharmacodynamic and clinical trial evidence for statin dose. Br J Clin Pharmacol. 2018;84(6):1128‐1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Maningat P, Gordon BR, Breslow JL. How do we improve patient compliance and adherence to long‐term statin therapy. Curr Atheroscler Rep. 2013;15(1):291‐301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Wolfe S. Rosuvastatin: winner in the statin wars, patients' health notwithstanding. BMJ. 2015;350(mar17 4):h1388. [DOI] [PubMed] [Google Scholar]
  • 11. Amarenco P, Bogousslavsky J, Callahan A, et al. High‐dose atorvastatin after stroke or transient ischemic attack. N Engl J Med. 2006;354:549‐559. [DOI] [PubMed] [Google Scholar]
  • 12. Meyer JH, Wilson AA, Sagrati S, et al. Serotonin transporter occupancy of five selected serotonin reuptake inhibitors at different doses: an [11C]DASB positron emission tomography study. Am J Psychiatry. 2004;161(5):826‐835. [DOI] [PubMed] [Google Scholar]
  • 13. ebmh.bmj.com/contents/suppl/2005/02/10/7.4.106.DC1/74106table.pdf
  • 14. Hannestad J, Gallezot J‐D, Planeta‐Wilson B, et al. Clinically relevant doses of methylphenidate significantly occupy the norepinephrine transporter in humans in vivo. Biol Psychiatry. 2010;68(9):854‐860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.file:///C:/Users/Simon/Documents/3%20Guidelines/ED50/Stimulants/ED50%20Dexamphetamine%20about%2010%20mg.pdf
  • 16. Boucher M, Duchene‐Marullaz P. Acebutol, metoprolol and propranolol in conscious dogs with chronic heart‐blocker: chronotropic effects and relation between depression of ventricular activity and beta‐adrenoceptor blocking potency. Br J Pharmac. 1980;70(2):335‐340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Heran BS, Wong MM, Heran IK, Wright JM. Blood pressure lowering efficacy of angiotensin converting enzyme (ACE) inhibitors for primary hypertension. Cochrane Database Syst Rev. 2008;(4):CD003823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Garcia D, Regan S, Crowther M, Hughes RA, Hylek EM. Warfarin maintenance dosing patterns in clinical practice. Implications for safer anticoagulation in the elderly population. Chest. 2005;127(6):2049‐2056. [DOI] [PubMed] [Google Scholar]
  • 19. Singh R. Does one size fit everyone? Replacement dose of levothyroxine in long‐standing primary hypothyroidism in adults. Indian J Endocrinol Metab. 2017;21(3):404‐409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Turgeon RD, Kolbert MR, Loewen P, Ellis U, McCormack JP. Higher versus lower doses of ACE inhibitors, angiotensin‐2 receptor blockers and beta‐blockers in heart failure with reduced ejection fraction: systematic review and meta‐analysis. PLoS One. 2019;14(2):e0212907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Dimmitt S, Stampfer H, Warren JB. Beta‐adrenoceptor blockers valuable but higher doses not necessary. Br J Clin Pharmacol. 2014;78(5):1076‐1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Ferner RE, Aronson JK. Susceptibility to adverse drug reactions. Br J Clin Pharmacol. 2019. 10.1111/bcp.14015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Blann AD, Fitzmaurice DA, Lip GYH. Anticoagulation in hospitals and general practice. Br Med J. 2003;326(7381):153‐156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Patel TK, Patel PB. Mortality among patients due to adverse drug reactions that lead to hospitalization: a meta‐analysis. Eur J Clin Pharm. 2018;74(6):819‐832. [DOI] [PubMed] [Google Scholar]
  • 25. Montane E, Arellano AL, Sanz Y, Roca J, Farre M. Drug‐related deaths in hospital inpatients: a retrospective cohort study. Br J Clin Pharmacol. 2018;84(3):542‐552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Moutlon JP, Mehta U, Parrish AG, et al. Mortality from adverse reactions in adult medical inpatients at four hospitals in South Africa: a cross‐sectional survey. Br J Clin Pharmacol. 2014;80:818‐826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Bollini P, Pampallona S, Tibaldi G, Kupelnick B, Munizza C. Effectiveness of antidepressants. Meta‐analysis of dose‐effect relationships in randomised clinical trials. Br J Psych. 1999;174:297‐303. [DOI] [PubMed] [Google Scholar]
  • 28. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/021992s004lbl.pdf
  • 29. Salvo F, Pariente A, Shakir S, et al. Sudden cardiac and sudden unexpected death related to antipsychotics: a meta‐analysis of observational studies. Clin Pharmacol Ther. 2016;99(3):306‐314. [DOI] [PubMed] [Google Scholar]
  • 30. Newcomer JW. Antipsychotic medications: metabolic and cardiovascular risk. J Clin Psychiatry. 2007;68(S4):8‐13. [PubMed] [Google Scholar]
  • 31. Ackenheil M, Weber K. Differing response to antipsychotic therapy in schizophrenia: pharmacogenomic aspects. Dialogues Clin Neurosci. 2004;6:71‐77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Macher J‐P, Corcq M‐A. Treatment goals: response and nonresponse. Dialogues Clin Neurosci. 2004;6:83‐91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Rink L, Braun C, Bschor T, Henssier J, Franklin J, Baethge C. Dose increase versus unchanged continuation of antidepressants after initial antidepressant treatment failure in patients with major depressive disorder: a systematic review and meta‐analysis of randomized double‐blind trials. J Clin Psychiatry. 2018;79:17r11693. [DOI] [PubMed] [Google Scholar]
  • 34. Davis JM, Chen NJ. Dose response and dose equivalence of antipsychotics. J Clin Psychopharmacol. 2004;24(2):192‐208. [DOI] [PubMed] [Google Scholar]
  • 35. DiBonaventura M, Gabriel S, Dupclay L, Gupta S, Kim E. A patient perspective of the impact of medication‐side effects on adherence: results of a cross‐sectional nationwide survey of patients with schizophrenia. BMC Psychiatry. 2012;12:20‐26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Reeve E, Gnijidic D, Long J, Hilmer S. A systematic review of the emerging definition of ‘deprescribing' with network analysis: implications for future research and clinical practice. Br J Clin Pharmacol. 2015;80(6):1254‐1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Tampi RR, Tampi DJ, Balachandran S, Srinivasan S. Antipsychotic use in dementia: a systematic review of benefits and risks from meta‐analyses. Ther Adv Chronic Dis. 2016;7(5):229‐245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Schneider LS, Tariot PN, Dagerman KS, et al. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer's disease. N Engl J Med. 2006;355(15):1525‐1538. [DOI] [PubMed] [Google Scholar]
  • 39. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia. Meta‐analysis of randomized placebo‐controlled trials. JAMA. 2005;294(15):1934‐1943. [DOI] [PubMed] [Google Scholar]
  • 40. Dimmitt SB, Stampfer HG. Low drug doses may improve outcomes in chronic disease. Med J Aust. 2009;191:511‐513. [DOI] [PubMed] [Google Scholar]
  • 41. Avorn J, Kesselheim A, Sarpatwari A. The FDA amendments act of 2007 – assessing its effects a decade later. N Engl J Med. 2018;379(12):1097‐1099. [DOI] [PubMed] [Google Scholar]
  • 42. https://www.fda.gov/RegulatoryInformation/LawsEnforcedbyFDA/SignificantAmendmentstotheFDCAct/FoodandDrugAdministrationAmendmentsActof2007/default.htm (cited 28 March 2019)
  • 43. https://www.ema.europa.eu/en/human‐regulatory/marketing‐authorisation/pharmacovigilance/risk‐management/risk‐management‐plans (cited 28 March 2019)
  • 44. Frau S, Pous MR, Luppino MR, Conforti A. Risk management plans: are they a tool for improving drug safety? Eur J Clin Pharmacol. 2010;66(8):785‐790. [DOI] [PubMed] [Google Scholar]
  • 45. Steinbuhl SR, Berger PB, Mann JT, et al. Early and sustained dual oral antiplatelet therapy following percutaneous coronary intervention. A randomized controlled trial. JAMA. 2002;288(19):2411‐2420. [DOI] [PubMed] [Google Scholar]
  • 46. Lucas CJ, Dimmitt SB, Martin JH. Optimising low dose methotrexate for rheumatoid arthritis – A Review. Br J Clin Pharmacol. 10.1111/bcp.14057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. van Echteld I, Wechalekar MD, Schlesinger N, Buchbinder R, Aletaha D. Colchicine for acute gout. Cochrane Database Syst Rev. 2014;(8):CD006190. [DOI] [PubMed] [Google Scholar]
  • 48. Ahern MJ, Reid C, Gordon TP, et al. Does colchicine work? The results of the first controlled study in acute gout. Aust N Z J Med. 1987;17(3):301‐304. [DOI] [PubMed] [Google Scholar]
  • 49. Terkeltaub RA, Furst DE, Bennett K, Kook KA, Crockett RS, Davis MW. High versus low dosing of oral colchicine for early acute gout flare: twenty‐four‐hour outcome of the first multicenter, randomized, double‐blind, placebo‐controlled, parallel group, dose‐comparison colchicine study. Arthritis Rheum. 2010;62(4):1060‐1068. [DOI] [PubMed] [Google Scholar]
  • 50. Andreasen F, Lauridsen IN, Hansent FA, Christensen S, Steiness E. Dose dependence of furosemide‐induced sodium excretion. J Pharmacol Exp Ther. 1989;248:1182‐1188. [PubMed] [Google Scholar]
  • 51. Felker GM, Lee KL, Bull DA, et al. Diuretic strategies in patients with acute decompensated heart failure. N Engl J Med. 2011;364(9):797‐805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Musini VM, Nazer M, Bassett K, Wright JM. Blood pressure‐lowering efficacy of monotherapy with thiazide diuretics for primary hypertension. Cochrane Database Syst Rev. 2014;(5):CD003824. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES