Skip to main content
Log in

Etidronic Acid

A Review of its Pharmacological Properties and Therapeutic Efficacy in Resorptive Bone Disease

  • Drug Evaluation
  • Published:
Drugs & Aging Aims and scope Submit manuscript

Summary

Synopsis

Etidronic acid is an orally and intravenously active bisphosphonate, which is believed to inhibit resorption of bone via a number of cellular mechanisms, including alteration of osteoclastic activity.

In studies of patients with symptomatic Paget’s disease, etidronic acid 5 to 20 mg/kg/day administered orally rapidly decreased the biochemical indices of bone turnover. Mineralisation defects informing bone may be avoided by the use of an initial dosage of 5 mg/kg/day for up to 6 months; dosages above 10 mg/kg/day should be limited to 3 months’duration, and dosages greater than 20 mg/kg/day should be avoided.

Although 3- day intravenous therapy with etidronic acid 7.5 mg/kg/day has shown superior efficacy to rehydration and forced diuresis in the management of hypercalcaemia of malignancy, the efficacy of the drug is lower than that of the newer bisphosphonates, pamidronic acid and clodronic acid.

Clinical studies involving postmenopausal women with established osteoporosis have indicated that oral etidronic acid 400 mg/day for 14 days as part of a 90- day cycle, repeated for up to 3 years, increases the bone mineral density (BMD) of the lumbar vertebrae and appears to reduce the incidence of vertebral fracture. Published data suggest that etidronic acid shows similar efficacy to hormone replacement therapy (HRT) in these respects. The above dosage also appears to be effective in preventing corticosteroid-induced osteoporosis when administered as part of an intermittent, cyclical regimen. Etidronic acid in higher dosages (10 to 20 mg/kg/day orally) is effective in reducing the incidence of heterotopic ossification and its ensuing complications in both neurological and post-surgical patients.

Etidronic acid is well tolerated by the majority of patients, with gastrointestinal complaints reported most commonly, but tends to delay the normal mineralisation of forming bone when administered continuously at higher dosages for prolonged periods. This is of little consequence where short term treatment is involved, but may be detrimental to those patients receiving longer courses of therapy. This effect may be minimised or avoided by using the lowest effective dosage for as short a time as possible (as in the above recommendations for Paget’s disease), or by the use of intermittent cyclical therapy (as in the management of osteoporosis).

Etidronic acid therefore retains a role in the management of resorptive bone disease, particularly in the treatment of Paget’s disease, the prevention of heterotopic ossification, and as a second-line option in postmenopausal osteoporosis. However, the development of newer bisphosphonate s requires that these compounds be continually compared and re-evaluated.

Pharmacodynamic Properties

Etidronic acid is adsorbed onto hydroxyapatite crystals in mineralised bone matrix, but it is currently thought that alterations in the number and activity of osteoclasts, together with other cellular mechanisms, are responsible for the anti-resorptive properties of this compound. It inhibits bone resorption both in vitro and in vivo, and appears to act preferentially on the axial skeleton. If administered continuously for prolonged periods at sufficiently high antiresorptive dosages, etidronic acid may impair the normal mineralisation of forming bone, as the dosage levels required to delay mineralisation are close to those used therapeutically. Recent data suggest, however, that this phenomenon may also be seen with the second generation bisphosphonate pamidronic acid at therapeutic dosages.

In patients with symptomatic Paget’s disease, etidronic acid decreases, or has no effect on, serum alkaline phosphatase (ALP) activity (a marker for osteoblastic activity), and serum calcium levels remain stable. Serum phosphate levels and renal tubular reabsorption of phosphate are increased, while serum creatinine, parathyroid hormone (PTH) and calcifediol (25-hydroxycholecalciferol) levels are generally unaffected.

In patients with hypercalcaemia of malignancy, etidronic acid reduces serum calcium levels when given intravenously at dosages of up to 500 mg/day for 3 to 7 days, the maximum effect being reached at approximately day 5. From the limited data available, serum creatinine levels and ALP activity appear unaffected, whereas urinary hydroxyproline (HDP) excretion (a marker of osteoclastic bone resorption) is reduced by approximately 50%.

Pharmacokinetic Properties

Etidronic acid is highly water soluble and its diffusion across lipid membranes is correspondingly poor. Oral bioavailability ranges from 1 to 10%. Between 20 and 50% of the orally absorbed dose becomes localised in bone, the remainder being excreted in the urine within 24 hours. The renal clearance of etidronic acid is 0.09 L/kg/h. The bisphosphonates have also been shown to accumulate in other tissues, including the stomach, liver, spleen and tracheal cartilage.

The plasma elimination half-life of etidronic acid is approximately 2 hours and the volume of distribution 0.3 to 1.3 L/kg. Etidronic acid has a high affinity for bone, where its elimination half-life exceeds 1 year. Animal studies with radiolabelled etidronic acid indicate that intermittent cyclic courses over 3 years would result in skeletal retention of 25 to 50 times the daily absorbed dose.

Etidronic acid has been shown to interfere with the skeletal binding of diagnostic radioisotopes such as technetium-labelled methylene diphosphonate (99Tcm-MDP).

Therapeutic Use

Sustained suppression of serum ALP activity and urinary HDP excretion to normal levels may be achieved in up to 76% of patients with Paget’s disease of bone after a single 6-month course of etidronic acid 5 to 20 mg/kg/day administered orally. These improvements may be maintained in some patients for up to 5 years. Subjective pain scores (bone, joint and mixed pain) are reduced to a clinically significant extent in a majority of patients, even in the presence of osteoarthritis, and these reductions persist for a similar period. No radiological evidence of improvement in skeletal structure is evident, however, after treatment with etidronic acid 5 to 20 mg/kg/day for periods of up to 5 years. Treatment of Paget’s disease with etidronic acid at the higher dosage range of 10 to 20 mg/kg/day has been associated with delayed mineralisation, indicated by osteoid seam widening, but these effects are not seen at the lower dosage of 5 mg/kg/day.

Data from a limited number of sequential studies indicate that oral etidronic acid 7 to 14 mg/kg/day for 6 months is superior to calcitonin ≤160 MRC units daily in reducing serum ALP levels and urinary HDP excretion in Paget’s disease. When etidronic acid is given together with synthetic human calcitonin, clinical and biochemical effects are similar to those of calcitonin alone. Clinical benefit may, however, be obtained more rapidly with combined treatment. Similar reductions in serum AP levels have been observed in patients with Paget’s disease after treatment with etidronic acid 5 to 20 mg/kg/day and clodronic acid 800 to 1600 mg/day.

Etidronic acid 7.5 mg/kg/day in combination with intravenous rehydration and furosemide 40 to 80 mg/day has shown superior efficacy to hydration + diuretic alone in the management of hypercalcaemia of malignancy. In patients resistant to rehydration, intravenous etidronic acid 7.5 mg/kg/day for 3 to 5 days produced normocalcaemia in 31 to 43% of patients. However, the drug was less effective in this respect, when given orally or intravenously, than single intravenous doses of pamidronic acid 30 to 60mg, clodronic acid 600mg or plicamycin 25 µg/kg, or intravenous gallium nitrate 200 mg/m /day for 5 days.

The majority of clinical studies involving etidronic acid in osteoporosis have involved postmenopausal women, with much interest being focused on cyclic or coherence (ADFR) regimens. Etidronic acid 400 mg/day orally for 14 days increased bone mineral density (BMD) of the lumbar vertebrae when given either as part of an ADFR regimen or cyclically without an activator. Treatment for 2 years has resulted in BMD increases versus baseline of up to 15.7%; this contrasts with unchanged or reduced BMD after placebo treatment or calcium supplementation alone. Published trials have also indicated a favourable effect of etidronic acid on vertebral fracture rate (VFR). Results of studies comparing etidronic acid with hormone replacement therapy (HRT) are equivocal. Recent data have suggested a preventative effect of cyclic courses of etidronic acid on corticoste-roid-induced osteoporosis.

Etidronic acid 10 to 20 mg/kg/day orally has been shown to be effective in reducing the incidence of heterotopic ossification and its ensuing complications in both neurological and post-surgical patients. Orthopaedic patients benefit from therapy initiated 4 to 6 weeks before surgery and continued for 3 months post-operatively; those with spinal cord injury require treatment within 60 days of trauma. Etidronic acid does not prevent ectopic bone formation in these patients, but delays its mineralisation, thus allowing patients to mobilise fully before heterotopic lesions are able to consolidate.

Tolerability

Clinical studies have indicated that etidronic acid is well tolerated by the majority of patients at dosages of up to 20 mg/kg/day. Gastrointestinal adverse events are most commonly reported in the literature, usually after oral therapy. Taste disturbance, often manifesting as a metallic taste sensation, has been reported by patients receiving intravenous etidronic acid for hypercalcaemia of malignancy and tumour-induced bone disease; normal taste perception returns rapidly after cessation of treatment.

No clinically significant abnormalities in hepatic function or serum biochemistry attributable to etidronic acid therapy have been documented. Renal tubular reabsorption of phosphate is increased, but this appears to be of no clinical consequence. Rare instances of renal failure have been attributed to etidronic acid given as high-dosage intravenous therapy for hypercalcaemia of malignancy (bolus or rapid infusion of up to 1g daily).

In patients with Paget’s disease undergoing treatment with etidronic acid, development of, or increase in, bone pain at pagetic sites has occurred in 1 to 10% of patients, the effect being dose-related and associated with adverse effects on bone mineralisation.

Histomorphometry has failed to demonstrate defective mineralisation, and desirable effects on bone remodelling have been observed, after up to 7 years’ cyclical treatment with oral etidronic acid 400 mg/day in women with established postmenopausal osteoporosis.

Dosage and Administration

Patients with Paget’s disease should be treated with an initial oral etidronic acid dosage of 5 mg/kg/day for up to 6 months. Dosages above 10 mg/kg/day should be reserved for patients in whom there is an overriding need to suppress bone turnover and should not exceed 3 months’ duration; it is inadvisable to give more than 20 mg/kg/day.

Intravenous dosages of 7.5 mg/kg/day following adequate rehydration are effective when given for 3 days in the management of malignant hypercalcaemia: the dose should be infused over at least 2 hours. Dosage reduction may be advisable in the presence of impaired renal function. Oral etidronic acid 20 mg/kg/day for up to 30 days may be used to maintain normocalcaemia, with the first dose administered on the day following the last intravenous dose.

Established vertebral osteoporosis may be treated with etidronic acid 5 to 10 mg/kg/day (usual dosage 400 mg/day) orally for 14 days, followed by a 76-day period of calcium supplementation, with this cycle repeated for up to 3 years.

For the prevention of heterotopic ossification following total hip replacement, etidronic acid 20 mg/kg/day should be given orally from 1 month before to 3 months after surgery, and spinal cord injury patients may benefit from 20 mg/kg/day for 2 weeks, followed by 10 mg/kg/day for a further 10 weeks. Treatment should start as soon as possible after injury.

Oral etidronic acid should not be administered with food or dairy products; mineral supplements and antacids will also impair gastrointestinal absorption and the drug should therefore be taken in the middle of a 4-hour fast. It may be taken as a single daily dose, although divided doses may be given if adverse gastrointestinal effects occur.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Hosking DJ. Advances in the management of Paget’s disease of bone. Drugs 1990; 40: 829–40

    PubMed  CAS  Google Scholar 

  2. Singer FR. Role of the biphosphonate etidronate in the therapy of cancer-related hypercalcemia. Semin Oncol 1990 Apr; 17 Suppl. 5: 34–9

    PubMed  CAS  Google Scholar 

  3. Papapoulos SE, Landman JO, Bijvoet OLM, et al. The use of bisphosphonates in the treatment of osteoporosis. Bone 1992; 13 Suppl. 1: S41–49

    PubMed  CAS  Google Scholar 

  4. Fleisch H. Bisphosphonates: pharmacology and use in the treatment of tumour-induced hypercalcaemic and metastatic bone disease. Drugs 1991; 42: 919–44

    PubMed  CAS  Google Scholar 

  5. Fleisch H. Bisphosphonates: mechanisms of action and clinical use. In: Mundy GR, Martin TJ, editors. Handbook of experimental pharmacology. Stuttgart, New York: Springer Verlag, 1993:377–418

    Google Scholar 

  6. Kaplan RA, Geho WB, Poindexter C, et al. Metabolic effects of diphosphonate in primary hyperparathyroidism. J Clin Pharmacol 1977; 17:410–9

    PubMed  CAS  Google Scholar 

  7. Bassani D, Sabatini M, Scanziani E, et al. Bone invasion by Walker 256 carcinoma, line A in young and adult rats: effects of etidronate. Oncology 1990; 47: 160–5

    PubMed  CAS  Google Scholar 

  8. Sahni M, Guenther HL, Fleisch H, et al. Bisphosphonates act on rat bone resorption through the mediation of osteoblasts. J Clin Invest 1993; 91: 2004–11

    PubMed  CAS  Google Scholar 

  9. Licata AA. From bathtub ring to osteorosis: a clinical review of the bisphosphonates. Cleve Clin J Med 1993; 60: 284–90

    PubMed  CAS  Google Scholar 

  10. Selby PL, Rehman MT, Economou G, et al. Cyclical etidronate therapy has a preferential effect on the axial skeleton in osteoporosis whatever the cause [abstract]. Summer meeting of the Bone and Tooth Society: marrow stromal cell differentiation — a conference in honour of Dr Maureen Owen; 1993 July 22–23: University Museum and Keble College, Oxford: 55

  11. Cosman E, Nieves J, Lindsay R. Comparative effects of cyclical etidronate and continuous estrogen on bone mass [abstract]. J Bone Miner Res 1993; 8 Suppl. 1: S260

    Google Scholar 

  12. Fleisch H, Russell RGG, Bisaz S, et al. The inhibitory effect of phosphonates on the formation of calcium phosphate crystals in vitro and on aortic and kidney calcification in vivo. Eur J Clin Invest 1970; 1: 12–8

    PubMed  CAS  Google Scholar 

  13. King WR, Francis MD, Michael WR. Effect of disodium ethane-1-hydroxy-1, 1 -diphosphonate on bone formation. Clin Orthop Relat Res 1971; 78: 251–70

    PubMed  CAS  Google Scholar 

  14. de Vries HR, Bijvoet OLM. Results of prolonged treatment of Paget’s disease of bone with disodium ethane-1 -hydroxy-1,1-diphosphonate (EHDP). Neth J Med 1974; 17: 281–98

    PubMed  Google Scholar 

  15. Adamson BB, Gallacher SJ, Byars J, et al. Mineralisation defects with pamidronate therapy for Paget’s disease. Lancet 1993; 342: 1459–60

    PubMed  CAS  Google Scholar 

  16. Krane SM. Paget’s disease of bone. In: Wilson JD, Braunwald E, Isselbacher KJ, et al., editors. Harrison’s principles of internal medicine, vol. 2. 12th ed. New York: McGraw-Hill, 1991: 1938–41

    Google Scholar 

  17. Devlin RD, Gutteridge DH, Prince RL, et al. Alterations in vitamin D metabolites during treatment of Paget’s disease of bone with calcitonin or etidronate. J Bone Miner Res 1990; 5(11): 1121–6

    PubMed  CAS  Google Scholar 

  18. Frame B, Marel GM. Paget disease: a review of current knowledge. Radiology 1981; 141: 21–4

    PubMed  CAS  Google Scholar 

  19. Seibel MJ, Cosman F, Shen V, et al. Urinary hydroxypyridinium crosslinks of collagen as markers of bone résorption and estrogen efficacy in postmenopausal osteoporosis. J Bone Miner Res 1993; 8: 881–9

    PubMed  CAS  Google Scholar 

  20. Hassager C, Jensen LT, Pødenphant J, et al. The carboxy-terminal pyridinoline cross-linked telopeptide of type I collagen in serum as a marker of bone résorption: the effect of nandrolone decanoate and hormone replacement therapy. Calcif Tissue Int 1994; 54: 30–3

    PubMed  CAS  Google Scholar 

  21. Campion GV, Delmas PD, Dieppe PA. Serum and synovial fluid osteocalcin (bone gla protein) levels in joint disease. Br J Rheumatol 1989; 28: 393–8

    PubMed  CAS  Google Scholar 

  22. Walton RJ, Russell RGG, Smith R. Changes in the renal and extrarenal handling of phosphate induced by disodium etidronate (EHDP) in man. Clin Sci Mol Med 1975; 49:45–56

    PubMed  CAS  Google Scholar 

  23. Boyce BF, Fogelman I, Ralston S, et al. Focal osteomalacia due to low-dose diphosphonate therapy in Paget’s disease. Lancet 1984; 1(8381): 821–4

    PubMed  CAS  Google Scholar 

  24. Norman DA, Zerwekh JE, Pak CYC. An apparent 1,25-dihydroxyvitamin D-independent stimulation of intestinal calcium absorption in patients with Paget disease of bone during a short-term diphosphonate therapy. Metabolism 1981; 30: 290–2

    PubMed  CAS  Google Scholar 

  25. Muir HG, Schabort I, Hough FS. Influence of disodium etidronate in Paget’s disease of bone. S Afr Med J 1987; 72: 470–2

    PubMed  CAS  Google Scholar 

  26. Gibbs CJ, Aaron JE, Peacock M. Osteomalacia in Paget’s disease treated with short term, high dose etidronate. BMJ 1986; 292: 1227–9

    PubMed  CAS  Google Scholar 

  27. Lawson-Matthew PJ, Guilland-Cumming DF, Yates AJP, et al. Contrasting effects of intravenous and oral etidronate on vitamin D metabolism in man. Clin Sci 1988; 74: 101–6

    PubMed  CAS  Google Scholar 

  28. Kanis JA, Urwin GH, Gray RE, et al. Effects of intravenous etidronate disodium on skeletal and calcium metabolism. Am J Med 1987; 82 Suppl. 2a: 55–70

    PubMed  CAS  Google Scholar 

  29. Jacobs TP, Gordon AC, Silverberg SJ, et al. Neoplastic hypercalcemia: physiologic response to intravenous etidronate disodium. Am J Med 1987; 82 Suppl. 2a: 42–50

    PubMed  CAS  Google Scholar 

  30. Mundy GR, Martin TJ. The hypercalcaemia of malignancy: pathogenesis and management. Metabolism 1982; 31(12): 1247–77

    PubMed  CAS  Google Scholar 

  31. Martin TJ, Suva LJ. Parathyroid hormone-related protein in hypercalcaemia of malignancy. Clin Endocrinol 1989; 31: 631–47

    CAS  Google Scholar 

  32. Recker RR, Saville PD. Intestinal absorption of disodium ethane-1-hydroxy-1,1-diphosphonate (disodium etidronate) using a deconvolution technique. Toxicol Appl Pharmacol 1973; 24: 580–9

    PubMed  CAS  Google Scholar 

  33. Mönkkönen J, Koponen H-M, Ylitalo P. Comparison of the distribution of three bisphosphonates in mice. Pharmacol Toxicol 1989; 65: 294–8

    Google Scholar 

  34. Larsson Å, Rohlin M. In vivo distribution of 14C-labeled ethylene-1-hydroxy-1, 1-diphosphonate in normal and treated young rats: an autoradiographic and ultrastructural study. Toxicol Appl Pharmacol 1980; 52: 391–9

    PubMed  CAS  Google Scholar 

  35. Mönkkönen J. A one year follow-up study of the distribution of 14C-clodronate in mice and rats. Pharmacol Toxicol 1988; 62:51–3

    PubMed  Google Scholar 

  36. Kasting GB, Francis MD. Retention of etidronate in human, dog, and rat. J Bone Miner Res 1992; 7: 513–22

    PubMed  CAS  Google Scholar 

  37. Geddes AD, Bevan JA, Kasting GB, et al. Retention of bisphosphonate in bone: quantitation of etidronate in iliaccrest biopsy samples from patients on intermittent therapy for postmenopausal osteoporosis. J Bone Miner Res 1992; 7 Suppl. 1: S328

    Google Scholar 

  38. Krasnow AZ, Collier BD, Isitman AT, et al. False-negative bone imaging due to etidronate disodium therapy. Clin Nucl Med 1988; 13: 264–7

    PubMed  CAS  Google Scholar 

  39. Chong WK, Cunningham DA. Intravenous etidronate as a cause of poor uptake on bone scanning, with a review of the literature. Clin Radiol 1991; 44: 268–70

    PubMed  CAS  Google Scholar 

  40. Demeo JH, Balseiro J, Cole TJ. Etidronate sodium therapy — a cause of poor skeletal radiopharmaceutical uptake. Semin Nucl Med 1991; 21: 332–4

    PubMed  CAS  Google Scholar 

  41. Hommeyer SH, Varney DM, Eary JF. Skeletal nonvisualisation in a bone scan secondary to intravenous etidronate therapy. J Nucl Med 1992; 33: 748–50

    PubMed  CAS  Google Scholar 

  42. Sandier ED, Parisi MT, Hattner RS. Duration of etidronate effect demonstrated by serial bone scintigraphy. J Nucl Med 1991; 32: 1782–4

    Google Scholar 

  43. Wallach S. Treatment of Paget’s disease. In: Advances in internal medicine, vol. 27. Chicago: Year Book Medical Publishers, 1982: 1–43

    Google Scholar 

  44. Merkow RL, Lane JM. Paget’s disease, part II. In: Sculco TP, editor. Orthopaedic care of the geriatric patient. St Louis: CV Mosby, 1985: 253–68

    Google Scholar 

  45. Siris ES, Canfield RE, Jacobs TP, et al. Long-term therapy of Paget’s disease of bone with EHDP. Arthritis Rheum 1980; 23: 1177–84

    PubMed  CAS  Google Scholar 

  46. Johnston Jr CC, Khairi MRA, Meunier PJ. Use of etidronate (EHDP) in Paget’s disease of bone. Arthritis Rheum 1980; 23: 1172–6

    PubMed  Google Scholar 

  47. Altman RD. Long-term follow-up of therapy with intermittent etidronate disodium in Paget’s disease of bone. Am J Med 1985; 79: 583–90

    PubMed  CAS  Google Scholar 

  48. Renier JC, Bontoux-Carre E, Leclere JM, et al. Treatment of Paget disease of bone: experience with etidronate [in French]. Sem Hop 1990; 66: 2535–45

    Google Scholar 

  49. Morales Piga A, Corres González J, Pérez Ruiz F. Long term results after ethanohydroxy diphosphonate treatment in Paget bone disease [in Spanish]. Rev Clin Esp 1991; 189: 213–7

    PubMed  CAS  Google Scholar 

  50. Morales Piga A, Garcia Vadillo A, Rodriguez Garcia A, et al. Ethylene-hydroxy-diphosphonate in the management of Paget’s disease of bone [in Spanish]. Rev Clin Esp 1985; 177: 273–7

    PubMed  CAS  Google Scholar 

  51. Bickerstaff DR, Douglas DL, Burke PH, et al. Improvement in the deformity of the face in Paget’s disease treated with diphosphonates. J Bone Joint Surg Br 1990; 72(1): 132–6

    PubMed  CAS  Google Scholar 

  52. Ravault A, Meunier PJ. Long-term follow-up of 88 patients with Paget’s disease treated by discontinuous courses of low-dose disodium etidronate [in French]. Rev Rheum Mal Osteoartic 1989; 56(4): 293–302

    CAS  Google Scholar 

  53. Dodd GW, Ibbertson HK, Fraser TRC, et al. Radiological assessment of Paget’s disease of bone after treatment with bisphosphonates EHDP and APD. Br J Radiol 1987; 60(717): 849–60

    PubMed  CAS  Google Scholar 

  54. Alexandre C, Chapuy MC, Bressot C, et al. Treatment of Paget’s disease of bone with ethane-1-hydroxy-1,1 -diphosphonate at low dosage. Nouv Presse Med 1980; 9: 3429–33

    PubMed  CAS  Google Scholar 

  55. Renier JC, Bontoux-Carre E, Bontoux L, et al. The treatment of Paget’s bone disease using ethane-1 hydroxy-1,1-diphosphonate (EHDP) [in French]. Revue du Rhumatisme 1982; 49: 87–92

    CAS  Google Scholar 

  56. Alexandre C, Meunier PJ, Edouard C, et al. Effects of ethane-1-hydroxy-1,1-diphosphonate (5 mg/kg/day dose) on quantitative bone histology in Paget’s disease of bone. Metab Bone Dis Rel Res 1981; 4 & 5: 309–16

    Google Scholar 

  57. Charhon S, Chapuy MC, Valentin-Opran A, et al. Intravenous etidronate for spinal cord dysfunction due to Paget’s disease [letter]. Lancet 1982; 1:391–2

    PubMed  CAS  Google Scholar 

  58. Renier JC, Delaby A, Bontoux-Carre E. A comparative study, in the same patients, of calcitonin and etidronate in Paget disease of bone [in French]. Sem Hop 1990; 66: 1433–7

    Google Scholar 

  59. PerryIII HM, Droke DM, Avioli LV. Alternate calcitonin and etidronate disodium therapy for Paget’s bone disease. Arch Intern Med 1984; 144: 929–33

    PubMed  Google Scholar 

  60. Jaeger P, Bischoff-Delaloye A, Burckhardt P. Le traitement combiné de la maladie de Paget de l’os par la calcitonine et le diphosphonate EHDP. Schweiz Med Wochenschr 1981; 111: 1893–7

    PubMed  CAS  Google Scholar 

  61. Evans RA. Treatment of Paget’s disease of bone. Med J Aust 1983; 1: 159–63

    PubMed  CAS  Google Scholar 

  62. Fraser TRC, Ibbertson HK, Holdaway IM, et al. Effective oral treatment of Paget’s disease of bone with APD (3-amino-l-hydroxypropylidene-1, 1 -bisphosphonate): a comparison with combined calcitonin + EHDP (l-hydroxyethylidene-1,1-bisphosphonate). Aust NZ J Med 1984; 14: 811–8

    CAS  Google Scholar 

  63. Dewis P, Prasad BK, Anderson DC, et al. Clinical experience with the use of two diphosphonates in the treatment of Paget’s disease. Ann Rheum Dis 1985; 44: 34–8

    PubMed  CAS  Google Scholar 

  64. Gray RES, Yates AJP, Preston CJ, et al. Duration of effect of oral diphosphonate therapy in Paget’s disease of bone. Q J Med 1987; 64: 755–67

    PubMed  CAS  Google Scholar 

  65. Preston CJ, Yates AJP, Beneton MNC, et al. Effective short term treatment of Paget’s disease with oral etidronate. BMJ 1986; 292: 79–80

    PubMed  CAS  Google Scholar 

  66. Warrell Jr RP. Questions about clinical trials in hypercalcaemia [editorial]. J Clin Oncol 1988; 6: 759–61

    PubMed  Google Scholar 

  67. Dodwell D, Howell A. Treating bony metastases: bisphosphonates look promising but their use should await the results of current studies. BMJ 1991; 303: 429–30

    PubMed  CAS  Google Scholar 

  68. Smith Jr JA. Palliation of painful bone metastases from prostate cancer using sodium etidronate: results of a randomized, prospective, double-blind, placebo-controlled study. J Urol 1989; 141(1): 85–7

    PubMed  Google Scholar 

  69. Schnur W. Relief of metastatic bone pain with etidronate disodium. Ohio State Med J 1987; 83: 62–5

    CAS  Google Scholar 

  70. Kawamura J, Tochigi H, Yanagawa M, et al. Effects of etidronate disodium (EHDP) on urogenital malignancies with bone metastasis: a multicentered collaborative evaluation [in Japanese]. Act Urol Jap 1988; 34: 528–37

    CAS  Google Scholar 

  71. Belch AR, Bergsagel DE, Wilson K, et al. Effect of daily etidronate on the osteolysis of multiple myeloma. J Clin Oncol 1991; 9: 1397–402

    PubMed  CAS  Google Scholar 

  72. Daragon A, Humez C, Michot C, et al. Treatment of multiple myeloma with etidronate: results of a multicentre double-blind study. Eur J Med 1993; 2: 449–52

    PubMed  CAS  Google Scholar 

  73. Ryzen E, Martodam RR, Troxell M, et al. Intravenous etidronate in the management of malignant hypercalcaemia. Arch Intern Med 1985; 145: 449–52

    PubMed  CAS  Google Scholar 

  74. Flores JF, Singer FR, Rude RK. Effectiveness of a 24-hour infusion of etidronate disodium in the treatment of hypercalcemia of malignant disease: a dose-ranging pilot study. Miner Electrolyte Metab 1991; 17: 390–5

    PubMed  CAS  Google Scholar 

  75. Hasling C, Charles P, Mosekilde L. Etidronate disodium for treating hypercalcaemia of malignancy: a double-blind, placebo-controlled study. Eur J Clin Invest 1986; 16: 433–7

    PubMed  CAS  Google Scholar 

  76. Singer FR, Ritch PS, Lad TE, et al. Treatment of hypercalcemia of malignancy with intravenous etidronate: a controlled, multicenter study. Arch Intern Med 1991; 151: 471–6

    PubMed  CAS  Google Scholar 

  77. Ralston SH, Gallacher SJ, Patel U, et al. Comparison of three intravenous bisphosphonates in cancer-associated hypercalcaemia [see comments]. Lancet 1989; 2(8673): 1180–2

    PubMed  CAS  Google Scholar 

  78. Gucalp R, Ritch P, Wiernik PH, et al. Comparative study of pamidronate disodium and etidronate disodium in the treatment of cancer-related hypercalcemia. J Clin Oncol 1992; 10: 132–42

    Google Scholar 

  79. Mundy GR, Wilkinson R, Heath DA. Comparative study of available medical therapy for hypercalcemia of malignancy. Am J Med 1983; 74: 421–32

    PubMed  CAS  Google Scholar 

  80. Warrell Jr JRP, Murphy WK, Schulman P. A randomized double-blind study of gallium nitrate compared with etidronate for acute control of cancer-related hypercalcemia. J Clin Oncol 1991; 9: 1467–75

    PubMed  Google Scholar 

  81. Schiller JH, Rasmussen P, Benson III A, et al. Maintenance etidronate in the prevention of malignancy-associated hypercalcemia. Arch Intern Med 1987; 147: 963–6

    PubMed  CAS  Google Scholar 

  82. Ringenberg QS, Ritch PS. Efficacy of oral administration of etidronate disodium in maintaining normal serum calcium levels in previously hypercalcemic cancer patients. Clin Ther 1987;9:318–25

    PubMed  CAS  Google Scholar 

  83. Rubin CD. Southwestern Internal Medicine Conference: age-related osteoporosis. Am J Med Sci 1991; 301: 281–98

    PubMed  CAS  Google Scholar 

  84. Ullrich I. Osteoporosis. West Virginia Med J 1991; 87: 347–50

    CAS  Google Scholar 

  85. Allen SH. Primary osteoporosis: methods to combat bone loss that accompanies aging. Postgrad Med 1993; 93: 43–55

    PubMed  CAS  Google Scholar 

  86. Turner CH. Toward a cure for osteoporosis: reversal of excessive bone fragility. Osteoporos Int 1991; 2: 12–9

    PubMed  CAS  Google Scholar 

  87. Stone MD, Hosking DJ. Treatment of osteoporosis: current and future. Ann Rheum Dis 1991; 50: 663–5

    PubMed  CAS  Google Scholar 

  88. Watts NB. Bisphosphonate therapy for postmenopausal osteoporosis. South Med J 1992 Aug; 85 Suppl. 2: 31–3

    Google Scholar 

  89. Fogelman I, Ryan P. Clinical experience with etidronate. J Obstet Gynaecol 1993; 13 Suppl. 1: S8–S10

    Google Scholar 

  90. Mallette LE, LeBlanc AD, Pool JL, et al. Cyclic therapy of osteoporosis with neutral phosphate and brief, high-dose pulses of etidronate. J Bone Miner Res 1989; 4(2): 143–8

    PubMed  CAS  Google Scholar 

  91. Silberstein EB, Schnur W. Cyclic oral phosphate and etidronate increase femoral and lumbar bone mineral density and reduce lumbar spine fracture rate over three years. J Nucl Med 1992; 33: 1–5

    PubMed  CAS  Google Scholar 

  92. Miller PD, Neal BJ, McIntyre DO, et al. Effect of cyclical therapy with phosphorus and etidronate on axial bone mineral density in postmenopausal osteoporotic women. Osteoporos Int 1991; 1: 171–6

    PubMed  CAS  Google Scholar 

  93. Kanis JA. What constitutes evidence for drug efficacy in osteoporosis? Drugs Aging 1993; 3: 391–9

    PubMed  CAS  Google Scholar 

  94. Marcus R. Cyclic etidronate: has the rose lost its bloom? Am J Med 1993; 95: 555–6

    PubMed  CAS  Google Scholar 

  95. Storm T, Thamsborg G, Steiniche T, et al. Effect of intermittent cyclical etidronate therapy on bone mass and fracture rate in women with postmenopausal osteoporosis. N Engl J Med 1990; 322: 1265–71

    PubMed  CAS  Google Scholar 

  96. Chaouat D, Belange G, Danan S, et al. One-year prospective study of disodium etidronate versus 17β estradiol for the prevention of postmenopausal osteoporosis. Rev Rhum Engl Ed 1993; 60: 311–5

    Google Scholar 

  97. Pacifici R, McMurtry C, Vered I, et al. Coherence therapy does not prevent axial bone loss in osteoporotic women: a preliminary comparative study. J Clin Endocrinol Metab 1988; 66: 747–53

    PubMed  CAS  Google Scholar 

  98. Watts NB, Harris ST, Genant HK, et al. Intermittent cyclical etidronate treatment of postmenopausal osteoporosis. N Engl J Med 1990; 323: 73–9

    PubMed  CAS  Google Scholar 

  99. Steiniche T, Hasling C, Charles P, et al. The effect of etidronate on trabecular bone remodeling in postmenopausal spinal osteoporosis: a randomized study comparing intermittent treatment and an ADFR regime. Bone 1991; 12(3): 155–63

    PubMed  CAS  Google Scholar 

  100. Evans RA, Somers NM, Dunstan CR, et al. The effect of low-dose cyclical etidronate and calcium on bone mass in early postmenopausal women. Osteoporos Int 1993; 3: 71–5

    PubMed  Google Scholar 

  101. Fujita T, Orimo H, Inoue T, et al. Double-blind multicenter comparative study with alfacalcidol of etidronate disodium (EHDP) in involutional osteoporosis [in Japanese]. Rinsho Hyoka 1993; 21: 261–302

    Google Scholar 

  102. Storm T, Thamsborg G, Sørensen HJ, et al. Long-term treatment with intermittent cyclical etidronate: effect on bone mass and fracture rate. J Bone Miner Res 1992; 7 Suppl. 1: S117

    Google Scholar 

  103. Harris ST, Watts NB, Jackson RD, et al. Four-year study of intermittent cyclic etidronate treatment of postmenopausal osteoporosis: three years of blinded therapy followed by one year of open therapy. Am J Med 1993; 95: 557–67

    PubMed  CAS  Google Scholar 

  104. Stark R, Civitelli R, Avioli L, et al. Comparison of etidronate therapy to calcium and estrogen therapy in postmenopausal women [abstract]. J Bone Miner Res 1992; 7 Suppl. 1: S329

    Google Scholar 

  105. Silverman SL, Greenwald M, Golde G. Frequency of non-responders to etidronate therapy [abstract]. J Bone Miner Res 1993; 8Suppl. 1: S342

    Google Scholar 

  106. Dambacher MA, Neff M, Schmitt S, et al. Etidronate in slow and fast loser osteoporotic patients [abstract]. 4th International Symposium on Osteoporosis and Consensus Development Conference; 1993 March 27–April 2: Hong Kong: 116

  107. Mulder H, Struys A. Intermittent cyclical etidronate in the prevention of corticosteroid-induced bone loss. Br J Rheumatol 1994; 33: 348–50

    PubMed  CAS  Google Scholar 

  108. Miller PD, Hamilos DL, Mclntyre DO, et al. Cyclical etidronate (CE) increases bone mass in patients with established steroid-induced osteoporosis (SIOP) [abstract]. 15th Annual Meeting of the American Society for Bone and Mineral Research; 1993 Sept 18–22: Tampa, Florida

  109. Stammen D, Worth H, Richter B, et al. Therapy of steroid-induced osteoporosis in adult asthmatics [abstract]. International Conference, American Lung Association and American Thoracic Society; 1991 May 12–15: Anaheim, California

  110. Skingle SJ, Crisp AJ. Etidronate prevents corticosteroid induced bone loss from lumbar spine but not from neck of femur [abstract]. Bone and Tooth Society, London. Winter Meeting; 1993 Dec 6: P4

  111. Adachi JD, Bensen WG, Bianchi F, et al. Intermittent cyclic etidronate therapy (ICT) prevents corticosteroid-induced bone loss [abstract]. American College of Rheumatology, 57th Annual Scientific Meeting; Nov 7–11: San Antonio, Texas

  112. Gallacher SJ, Anderson K, Fenner JAK, et al. A comparison of intermittent oral etidronate and intravenous pamidronate in the treatment of corticosteroid-associated osteoporosis [abstract]. 23rd European Symposium on Calcified Tissues; 1993 April 25–29: Heidelberg

  113. Subbarao JV. Pseudoarthrosis in heterotopic ossification in spinal cord-injured patients. Am J Phys Med Rehab 1990; 69(2): 88–90

    CAS  Google Scholar 

  114. Nollen JG. Effects of ethylhydroxydiphosphonate (EHDP) on heterotopic ossification. Acta Orthop Scand 1986; 57: 358–61

    PubMed  CAS  Google Scholar 

  115. Stover SL, Hahn HR, Miller JM. Disodium etidronate in the prevention of heterotopic ossification following spinal cord injury (preliminary report). Paraplegia 1976; 14: 146–56

    PubMed  CAS  Google Scholar 

  116. Bijvoet OLM, Nollen AJG, Slooff TJJH, et al. Effect of a diphosphonate on para-articular ossification after total hip replacement. Acta Orthop Scand 1974; 45: 926–34

    PubMed  CAS  Google Scholar 

  117. Rech M. The effects of etidronate disodium in the prevention of heterotopic ossification following hemiarthroplasty of the hip: continuing studies. Contemp Orthop 1986; 13: 31–8

    Google Scholar 

  118. Finerman GAM, Stover SL. Heterotopic ossification following hip replacement or spinal cord injury: two clinical studies with EHDP. Metab Bone Dis Rel Res 1981; 4 & 5: 337–42

    Google Scholar 

  119. Reginster JY, Gritten C, Diverse P, et al. Treatment of Paget’s disease with low-dose etidronate disodium (5 mg/kg/day). Appearance of transitory reversible leukopenia [in French]. Rev Rhum 1985; 52: 145–50

    PubMed  CAS  Google Scholar 

  120. Bounameaux HM, Schifferli J, Montani J-P, et al. Renal failure associated with intravenous diphosphonates [letter]. Lancet 1983; 1:471

    PubMed  CAS  Google Scholar 

  121. Kanis JA, Preston CJ, Yates AJP, et al. Effects of intravenous diphosphonates on renal function [letter]. Lancet 1983; 1: 1328

    PubMed  CAS  Google Scholar 

  122. Eyres KS, Marshall P, McCloskey E. Spontaneous fractures in a patient treated with low doses of etidronic acid (disodium etidronate). Drug Saf 1992; 7: 162–5

    PubMed  CAS  Google Scholar 

  123. Nagant de Deuxchaines C, Dufour JP, Devogelaer JP, et al. Etidronate and the risk of fractures [letter]. Lancet 1985; 1:610–1

    Google Scholar 

  124. Steiniche T, Storm T, Thamsborg G, et al. A histomorphometric evaluation of trabecular bone in osteoporotic patients on long term etidronate treatment. J Bone Miner Res 1992; 7 Suppl. 1:S330

    Google Scholar 

  125. Storm T, Steiniche T, Thamsborg G, et al. Changes in bone histomorphometry after long-term treatment with intermittent, cyclic etidronate for postmenopausal osteoporosis. J Bone Miner Res 1993; 8: 199–208

    PubMed  CAS  Google Scholar 

  126. Ott SM, Woodson GC, Huffer WE, et al. Bone histomorphometric changes after cyclical therapy with phosphate and etidronate disodium in women with postmenopausal osteoporosis. J Clin Endocrinol Metab 1994; 78: 968–72

    PubMed  CAS  Google Scholar 

  127. Alexandre C, Tavan P, Chappard D, et al. Intermittent administration of etidronate (Didronel®) in involutional osteoporosis. In: Christiansen C, Overgaard K, editors. Osteoporosis 1990, 3. 3rd International Symposium on Osteoporosis; 1990 Oct 14–20: Copenhagen. Aalborg: Handelstrykkeriet Aalborg, 1990: 1441–3

    Google Scholar 

  128. Gallacher SJ, Boyle IT, Capell HA. Pseudogout associated with the use of cyclical etidronate therapy. Scott Med J 1991; 36:49

    PubMed  CAS  Google Scholar 

  129. Bockman RS. Etidronate for postmenopausal osteoporosis [letter]. N Engl J Med 1990; 323: 1209–10

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Additional information

Various sections of the manuscript reviewed by:H. Fleisch, Department of Pathophysiology, University of Berne, Berne, Switzerland; S.T. Harris, Departments of Medicine and Radiology, University of California, San Francisco, California, USA; C. Hasling, Department of Internal Medicine, Odder Centralsygehus, Odder, Denmark; J.A. Kanis, Sheffield Metabolic Bone Unit, University of Sheffield, Sheffield, England; A.A. Licata, Cleveland Clinic Foundation, Cleveland, Ohio, USA; D.E. Meier, Department of Geriatrics, Mount Sinai Medical Center, New York, New York, USA; S.H. Ralston, Department of Medicine and Therapeutics, University of Aberdeen, Aberdeen, Scotland; J.C. Renier, Service de Rhumatologie, Centre Hospitalier Régional et Universitaire, Angers, France; R.G.G. Russell, Department of Human Metabolism and Clinical Biochemistry, University of Sheffield, Sheffield, England; F.R. Singer, John Wayne Cancer Institute, Santa Monica, California, USA; N.B. Watts, Section of Internal Medicine, Emory Clinic, Atlanta, Georgia, USA; C. Wüster, Department of Internal Medicine I, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Dunn, C.J., Fitton, A. & Sorkin, E.M. Etidronic Acid. Drugs & Aging 5, 446–474 (1994). https://doi.org/10.2165/00002512-199405060-00006

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00002512-199405060-00006

Keywords

Navigation