Scolaris Content Display Scolaris Content Display

Bifosfonatos para el cáncer de próstata avanzado

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

Referencias de los estudios incluidos en esta revisión

Adami 1989 {published data only}

Adami S, Mian M. Clodronate therapy of metastatic bone disease in patients with prostatic carcinoma. Recent Results in Cancer Research 116: 67‐72, 1989;116:67‐72.

Dearnaley 2003 {published data only}

Dearnaley DP, Sydes MR, Mason MD, Stott M, Powell CS, Robinson ACR, Thompson PM, Moffat LE, Naylor SL, Parmar MKB. A double‐blind, placebo‐controlled, randomised trial of oral sodium clodronate for metastatic prostate cancer (MRC PRO5 Trial). Journal of the National Cancer Institute, vol 95, No 17 Sep 3, 1300‐11, 2003;95(No 17):1300‐1311.

Elomma 1992 {published data only}

Elomma I, Kylmata T, Tammela T, Vitanen J, Ottelin M, Ruutu K, Jauhiainen M, Ala‐Opas M, Roos L, Seppanen J, Alfthan O. Effect of oral clodronate on bone pain: A controlled study in patients with metastatic prostate cancer.. International Journal of Urology and Nephrology 1992;24 (2):159‐166.

Ernst 2003 {published data only}

Ernst DS, Tannock IF, Winquist EW, Venner PM, Reyno L, Moore MJ, Chik, Ding K, Elliot C, Parulekar W. Randomized, double‐blind, controlled trial of mitoxantron/prednisone and clodronate versus mitoxantrone/prednisone and placebo in patiens with hormone‐refractory prostate cancer and pain. Journal of Clinical Oncology 2003;21(Sept 1 (17)):3335‐3342.

Kylmala 1993 {published data only}

Kylmala T, Tammela T, Risteli L, Risteli J, Taube T, Elomma I. Evaluation of the effect of oral clodronate on skeletal metastases with type I collagen metabolites. A controlled trial of the Finnish Prostate Cancer Group. European Journal of Cancer 1993;29A(6):821‐825.

Kylmala 1997 {published data only}

Kylmala T, Taube T, Tammela TL. Concomitant i.v. and oral clodronate in the relief of bone pain: A double‐blind placebo‐controlled study in patients with metastatic prostate cancer. British Journal of Cancer 1997;76:939‐942.

Saad 2002 {published data only}

Saad F, Gleason DM, Murray R. A randomized, placebo‐controlled trial of zoledronic acid in patients with hormone‐refractory metastatic prostate carcinoma. Journal of the National Cancer Institute 94: 1458‐68, 2002;94(No 19):1458‐1468.

Small 2003 {published data only}

Small EJ, Matthew RS, Seaman JJ, Petrone S, Kowalski MO. Combined analysis of two multicenter, randomized, placebo‐controlled studies of pamidronate disodium for the palliation of bone pain in men with metastatic prostatic cancer. Journal of Clinical Oncology 2003;21(No 23 (Dec1)):4277‐4284.

Smith 1989 {published data only}

Smith JA. Palliation of painful bone metastases from prostate cancer using sodium etidronate: results of a randomized, prospective, double‐blind, placebo‐controlled study. Journal of Urology 1989;141:85‐87.

Strang 1997 {published data only}

Strang P, Nilsson S, Brandstedt S. The analgesic efficacy of clodronate compared with placebo inpatients with painful bone metastases from prostatic cancer. Anticancer Research 1997;17:4717‐4721.

Referencias de los estudios excluidos de esta revisión

Adami 1985 {published data only}

Adami S, Salvagno G, Guarrera G. Dichloromethylene‐diphosphonate in patients with prostatic carcinoma metastatic to the skeleton. Journal of Urology 1985;134:1152‐1154.

Carey 1988 {published data only}

Carey PO, Lippert MC. Treatment of painful prostatic bone metastases with oral etidronate disodium. Urology 1988;32:403‐407.

Clarke 1991 {published data only}

Clarke NW, Holbrook IB, McClure J, George NJ. Osteoclast inhibition by pamidronate in metastatic prostate cancer: a preliminary study. British Journal of Cancer 1991;63(3):420‐423.

Cresswell 1995 {published data only}

Cresswell SM, English PJ, Hall RR. Pain relief and quality‐of‐life assessment following intravenous and oral clodronate in hormone‐escaped metastatic prostate cancer. British Journal of Urology 1995;76:360‐365.

Fernandez‐Conde 1997 {published data only}

Fernandez‐Conde M, Alcover J, Aaron JE, Ordi J, Carretero P. Skeletal response to clodronate in prostate cancer with bone metastases.. American Journal of Clincial Oncology 1997;20(5):471‐476.

Heidenreich 2001 {published data only}

Heidenreich A, Hoffmann R, Engelmann. The use of bisphosphonate for the palliative treatment of painful bone metastasis due to hormone refractory prostate cancer. Journal of Urology 2001;165(1):136‐140.

Heidenreich 2002 {published data only}

Heidenreich A, Elert A, Hofmann R. Ibandronate in the treatment of prostate cancer associated painful osseous metastases. Prostate Cancer and Prostatic Diseases 2002;5(3):231‐235.

Jagdev 2001 {published data only}

Jagdev SP, Purohit OP, Heatley S, Herling C, Coleman RE. Comparison of the effects of intravenous pamidronate and oral clodronate on symptoms and bone resorption in patients with metastatic bone disease. Annals of Oncology 2001;12:1433‐1438.

Kylmala 1994 {published data only}

Kylmala T, Tammela TL, Lindholm TS. The effect of combined intravenous and oral clodronate treatment on bone pain in patients with metastatic prostate cancer. Annales Chirurgiae et Gynaecologiae 1994;83(4):316‐319.

Magnusson 1998 {published data only}

Magnusson P, Larssson L, Englund G, Larsson B, Strang P, Selin‐Sjogren L. Differences of bone alkaline phosphatase isoforms in metastatic bone disease and discrepant effects of clodronate on different skeletal sites indicated by location of pain. Clinical Chemistry 1998;44(8 Pt 1):1621‐1628.

Pelger 1998 {published data only}

Pelger RC, Hamdy NA, Zwinderman AH. Effects of the bisphosphonate olpadronate in patients with carcinoma of prostate metastatic to the skeleton. Bone 1998;22:403‐408.

Taube 1994 {published data only}

Taube T, Kylmala T, Lamberg‐Allardt C. The effect of clodronate on bone in metastatic prostate cancer: Histomorphonmetric report of a double‐blind randomised placebo‐controlled study. European Journal of Cancer 1994;30:751‐758.

Vorreuther 1992 {published data only}

Vorreuther R, Klotz T, Engelking R. Clodronate in the palliative therapy of bone‐metastasized prostate carcinoma. Urologe A 1992;31(2):63‐66.

Vorreuther 1993 {published data only}

Vorreuther R. Bisphosphonates as an adjunct to palliative therapy of bone metastases from prostate carcinoma. A pilot study on clodronate.. British Journal of Urology 1993;72 (5 Pt 2):792‐795.

Adami 1997

Adami S. Bisphosphonates in prostate cancer. Cancer 1997;80(S8):1674‐1679.

Begg 1996

Begg C, Cho M, Eastwood S, Horton R, Moher D, Olkin I, Pitkin R, Rennie D, Schulz KF, Simel D, Stroup KF. Improving the quality of reporting of randomised controlled trials. The CONSORT statement. The Journal of the American Medical Association 1996;276(8):637‐639.

Bubendorf 2000

Bubendorf L, Schopfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC, Mihatsch MJ. Metastatic patterns of prostate cancer: an autopsy study of 1589 patients. Human Pathology 2000;31(5):578‐583.

Campbell 2004

Campbell MK, Elbourne DR, Altman DG for the CONSORT Group. CONSORT statement: extension to cluster randomised trials. British Medical Journal 2004;328(7441):702‐708.

Carlin 2000

Carlin BI, Andriole GL. The natural history, skeletal complications, and management of bone metastases in patients with prostate carcinoma. Cancer 2000;88(12 suppl):2989‐2994.

Clohisy 2002

Clohisy DR, Mantyh PW. Bone Cancer Pain. Cancer 2002;97(3 suppl):866‐873.

Cranney 2002

Cranney A, Gyatt G, Griffith L, Tugweel P, Rosen C. Meta‐analyses of therapies for postmenopausal osteoporosis. IX: Summary of meta‐analyses of therapies for postmenopausal osteoporosis. Endocrine Reviews 2002;23(4):570‐578.

Devogelaer 2000

Devogelaer D. Treatment of bone diseases with bisphosphonates, excluding osteoporosis. Current Opinion in Rheumatology 2000;12:331‐335.

Djulbegovic 2004

Djulbegovic B, Wheatley K, Ross J, Clark O, Bos G, Goldschmidt H, Cremer F, Alsina M, Glasmacher A. Bisphosphonates in multiple myeloma. The Cochrane Database of Systematic Reviews 2002;Issue 4:Art. No CD 003188.

Eaton 2003

Eaton CL, Colby L, Coleman RE. Pathophysiology of bone metastases from prostate cancer and the role of bisphosphonates in treatment. Cancer Treatment Reviews 2003;29(3):189‐198.

Fleisch 2002

Fleisch H. Development of bisphosphonates. Breast Cancer Research 2002;4:30‐34.

Frydenberg 1997

Frydenberg M, Stricker PD, Kaye KW. Prostate cancer diagnosis and treatment. Lancet 1997;349:1681‐1687.

Goodin 2002

Goodin S, Rao KV, DiPaola RS. State‐of‐the‐art treatment of metastatic hormone‐refractory prostate cancer. The Oncologist 2002;7:360‐370.

Hollis 1999

Hollis S, Campbell F. What is meant by intention to treat analysis? Survey of published randomised controlled trials. British Medical Journal 1999;319(7211):670‐674.

Jadad 1998

Jadad AR, Moher D, Klassen TP. Guides for reading and interpreting systematic reviews: II. How did the authors find the studies and assess their qualtiy?. Archives of Pediatrics and Adolescent Medicine 1998;152(8):812‐817.

Juni 2001

Juni P, Altman DG, Egger M. Systematic reviews in health care: assessing the quality of controlled clinical trials. British Medical Journal 2001;323(7303):42‐46.

Moher 1998

Moher D, Jadad AR, Klassen TP. Guides for reading and interpreting systematic reviews: III. How did the authors synthesize the data and make their conclusions?. Archives of Pediatrics and Adolescent Medicine 1998;152(9):915‐920.

Moher 1999

Moher D, Cook DJ, Jadad AR, Tugwell P, Moher M, Jones A, Pham B, KlassenTP. Assessing the quality of reports of randomised trials: implications for the conduct of meta‐analyses. Health Technology Assessment1999; Vol. 3, issue 12.

Morris 2003

Morris MJ, Scher HI. Cinical approaches to osseous metastases in prostate cancer. The Oncologist 2003;8:161‐173.

Pavlakis 2005

Pavlakis N, Schmidt M, Stockler M. Bisphosphonates for breast cancer. Cochrane Database of Systematic Reviews 2005, Issue 3, Art No. CD003474.

Reid 2003

Reid IR. Bisphosphonates: new indications and methods of administration. Current Opinion in Rheumatology 2003;15:458‐463.

Saad 2004

Saad F, Schulman CC. Role of bisphosphonates in prostate cancer. European Urology 2004;45:26‐34.

Santini 2003

Santini D, Gentilucci V, Vincenzi B, Picardi A, Vasaturo F, La Cesa A, Onori N, Scarpa S, Tonini G. The antineoplastic role of bisphosphonates: from basic reasearch to clinical evidence. Annals of Oncology 2003;14:1468‐1476.

Saunders 2004

Saunders Y, Ross JR, Broadley KE, Edmonds PM, Patel S. Systematic review of bisphosphonates for hypercalcaemia of malignancy. Palliative Medicine 2004;18(5):418‐431.

Turk 2003

Turk DC, Dworkin RH, Allen RR, Bellamy N, Brandenburg N, Carr DB, Cleeland C, Dionne R, Farrar JT, Galer BS, Hewitt DJ, Jadad AR, Katz NP, Kramer LD, Manning DC, McCormick CG, McDermott MP, McGrath P, Quessy S, Rappaport BA, Robinson JP, Royal MA, Simon L, Stauffer JW, Stein W, Tollett J. Witter J. Core outcome domains for chronic pain clinical trials: IMMPACT recommendations. Pain 2003;106:337‐345.

Wong 2004

Wong R, Wiffen PJ. Bisphosphonates for the relief of pain secondary to bone metastases. Cochrane Database of Systematic Reviews 2002, Issue 2, Art No. CD002068.

Referencias de otras versiones publicadas de esta revisión

Adami 1997

Adami S. Bisphosphonates in prostate carcinoma. Cancer 1997;80(S8):1674‐1679.

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Adami 1989

Methods

Three randomized single‐blinded controlled studies
Quality score (1/0/0)
Outcomes: pain and analgesic consumption
Pain assessment tool: 20cm VAS
Pain response definition: not specified
Analgesic consumption definition: daily consumption of analgesic drugs (ketoprofen)
Pain assessment schedule: 0,1,2,4 week.

Participants

Prostatic carcinoma. Bone metastasis (radiographic). Pain requirement: not specified.
Cointerventions: orchidectomy at least 8 months before study (n=16), extramustine but disease refractory (n=16), distribution not specified.
Randomized 56 patients.
Baseline pain (mean 20cm VAS): active 16/20, control 12/20

Interventions

Study 1: Placebo (n=6) versus Clodronate i.v. infusion 300mg /day x 2 weeks (n=7)
Study 2: Clodronate oral 1200mg/day x 2 weeks (n=11) versus Clodronate i.m. 100mg/day x 2 weeks (n=12)
Study 3: Clodronate i.v. x 2 weeks then stop (n=13) versus Clodronate i.v. x 2 weeks then clodronate oral 1200mg/day x at least 6 weeks (n=18)

Outcomes

Study 1: Mean pain score and analgesic consumption at 0,1,2,4 week. No S.D. provided.
Study 2: Mean pain score and analgesic consumption at 0,1,2,4 week. No S.D. provided.
Study 3: Mean percentage change in pain score at 1,2,4,6,8 week. No S.D. provided.
No haematological toxicity reported.

Notes

Dearnaley 2003

Methods

Randomized double‐blind placebo‐controlled study
Quality score (2/2/1)
Primary outcome: symptomatic bone progression‐free survival (osseous disease requiring increase analgesic, radiotherapy, change in hormonal therapy, pathological fracture or spinal cord compression)
Secondary outcomes: overall survival, toxicity, bone events, type of progressive disease, analgesic consumption, WHO performance status, PSA, alkaline phosphatase levels.
Pain assessment criteria: nil
Definition of pain response:
Analgesic: time to first regular use of analgesics of any strength.
Follow‐up schedule: 4 week, 3,6,9,12,15,18,21,24 months, then every 6 months (median follow up 59 months).

Participants

Prostate cancer bone metastases.
Pain not required.
Commencing or responding to hormonal therapy; no previous use of long‐term hormone therapy; normocalcaemia; WHO performance status 2 or less; bisphosphonates naive; serum creatinine less than 2 x ULN; no other active malignancy; no acute severe bowel inflammatory conditions; no serious concomitant disease; no drug trial within 12 months.
Randomized 311 patients: active 155, control 156, analysed active 155, control 156.

Interventions

Active: Clodronte oral 2080mg/day up to 3 years
Control: Placebo up to 3 years

Outcomes

Longer time to first regular use of analgesics in active group, HR = 1.12 (95%CI = 0.86‐1.45, P=0.41, data not shown)
Longer Symptomatic BPFS in active group, HR = 0.79 (0.61‐1.02, p.066)
Number of SBP events: active 94/155, control 103/156
Any death: active 111/155, control 124/156
Prostate cancer death: active 91/155 control 105/156
Bone progression as first event: active 89/155, control 98/156.
Disease progression: 106/155, 112/156.
Patients with adverse effects: active 78/155, control 53/156
Bone disease progression: 86/155, 98/156
Higher risk of adverse effects in active group, HR = 1.71 (1.21‐1.41, P 0.002),
Number of adverse events: Total active 123/155, control 82/156, including gastrointestinal (active 31/155 control 21/156), cardiovascular, joint pain, dyspnoea, urinary, skin, second primary malignancy, tiredness, central nervous system, bone pain/fracture, sweats, dizziness, headaches, abnormal blood counts (active 2/155 control 3/156) or biochemistry, paresthesia, sleep, hepatic, memory, gastrointestinal bleeding, etc.
Change in WHO performance status (HR 0.71, CI=0.56‐0.92, p.008).
Sustained deterioration of WHO performance status: active 79/155 control 98/156
Worsened WHO performance status, HR = 0.71, 95%CI = 0.56‐0.92, P=0.008, favours active.
Improved WHO performance before reaching primary endpoint: active 22/155, control 17/156.
Estimated median PSA during first 2 years: active 5.0, control 10.4 (P=0.053, data not shown).

Notes

Elomma 1992

Methods

Randomized placebo‐controlled parallel study. Blinding not mentioned.
Quality score 1/0/1
Primary outcome: pain
Secondary outcomes: performance status, bone pain, analgesic use, serum biochemical markers, side effects.
Pain assessment criteria: presence or absence of bone pain evaluated by patient and doctor
Pain response definition: no pain
Analgesic consumption assessment: not specified.
Analgesic response definition: no analgesic.
Pain assessment schedule: 0,1,3,6 month.

Participants

Prostate cancer refractory to at least one hormonal therapy. Bone metastases required. Pain required (with daily use of analgesics).
Life expectancy at least 3 months.
RT not allowed.
Randomized 75 patients: active 36, placebo 39

Interventions

Active arm: Clodronate oral 3.2g/day for 1 month then 1.6g/day for 5 months. Total 6 months.
Control arm: Placebo
Cointervention: All patient received estramustine 280mg bid

Outcomes

No pain (doctor evaluation) at 0, 1, 3, 6 months: clodronate 0/36 9/29 5/22 3/11; placebo 0/39 5/34 1/24 1/13.
No pain (patient evaluation) at 0, 1, 3, 6 months: clodronate 0/36 10/29 6/21 2/11; placebo 0/39 6/34 1/23 2/13
No analgesic at 0, 1, 3, 6 months: clodronate 0/36 11/29 5/22 2/11; placebo 0/39 6/34 3/24 3/13
Median survival and survival rates: no difference
All death: active 12/36, control 10/39.
Cancer death: active 9/36, control 6/39.
Performance status: ambulatory rate at 1, 3 month: active 30%, 40%, control 20%, 40%
Biochemical markers and blood count: no difference
Patients discontinued trial at 1,3,6 months: active 2,3,1, control 3,4,2.
Side effects uncommon.

Notes

Ernst 2003

Methods

Randomized double‐blind controlled study
Quality score (1/2/1)
Primary outcome: palliative response, defined as either 2‐point reduction in PPI (present pain intensity score, six‐point, 0‐5) or >50% decrease in analgesic score.
Secondary outcomes: time to symptomatic progression, duration of palliative response, PSA response (>50% decrease from baseline), morbid skeletal events (hypercalcaemia, pathologic fractures, palliative radiotherapy), toxicity, HRQOL. Urine biochemical markers.
Pain assessment: six‐point PPI scale 0‐5 (Present pain intensity scale of the McGill‐Melzack Pain Questionnaire)
Analgesic assessment: Analgesic score defined by total number of analgesics units (1 unit = standard doses of non‐opioid, 2 units = morphine 10‐mg equivalents).
Pain assessment schedule: every 3 week.

Participants

Prostate cancer. Progressive bone disease. Castration testosterone levels. Hormonal treatment stopped before randomization.
Pain required: PPI = 1 or more. Stable analgesic intake required.
ECOG performance status < 3, adequate baseline cardiac, haematologic and biochemical function, able to complete pain and QOL scores.
Exclusion: prior malignancy, more than one previous chemotherapy regimen or a previous regimen containing mitoxantrone or an anthracycline, precious bisphosphonate therapy, treatment with radiotherapy within the previous 4 weeks or radioisotopes within the previous 8 weeks, radicular or back pain suggestive of epidural metastases, potential spinal cord or nerve root compression, impending pathological fracture, and uncontrolled cardiac failure or active infection.
Randomized 227 patients: active 115, control 112; ineligible: active 11, control 7; analysed: active 104, control 105.
Baseline pain, PPI 1‐2, active 78/104, control 82/105; PPI 3‐4, active 26/104, control 23/105.

Interventions

Active arm (n=115): clodronate 1500mg i.v. q3w until disease progression in responding patients.
Control arm(n=112): placebo q3w
Cointervention: mitoxantrone 12mg/m2 iv q3w and prednisolone 5mg po bid.

Outcomes

Palliative response: active 49/115, control 41/112.
Pain response: active 34/115 control 27/112.
Analgesic response: active 34/115 control 32/112.
No need for analgesic: active 33/115 control 27/112.
Any death: active 87/115 control 89/112. All except one caused by disease progression.
PSA response: active 30/115 control 30/112
Disease progression: 58/115 control 68/112
Requirement of local radiotherapy: active 18/115 control 16/112
Morbid skeletal events: results not reported.
Adverse effects (grade3/4) (active:control): granulocytopenia 14/115 14/112, anaemia 8/115 5/112, thrombocytopenia 2/115 4/112, cardiovascular 0/115 3/112 nausea/vomiting 9/115 7/112 headache 4/115 1/112 dyspnoea 4/115 7/112 infection 7/115 3/112.
QOL response: active 39/115 control 44/112
Withdraws: active 11/115, 7/112

Notes

Kylmala 1993

Methods

Randomized controlled study
Quality score (1/0/1)
Primary outcome: type I collagen metabolites.
Other outcomes: pain, analgesic use, bone scan changes, serum calcium, phosphate, acid phosphatase, PSA, creatinine, transaminase.
Pain assessment criteria: presence or absence of bone pain
Definition of pain response: absence of pain.
Analgesic measurement: with or without analgesic
Pain assessment schedule: not stated, diagrams suggests this followed the assessment schedule for other parameters i.e. 0,1,3,6 months

Participants

Prostate cancer, hormone refractory.
Bone metastases required. Pain required. Analgesic consumption required.
Radiotherapy not allowed in preceding 2 months and during trial.
Life expectancy at least 3 months.
Randomized 99 patients: active 50, placebo 49.

Interventions

Active arm: oral clodronate 3.2g/day x 1 month then 1.6g/day x 5 months; extramustine 280mg twice daily
Control arm: open control with extramustine 280mg twice daily

Outcomes

Percentage of patient free from bone pain: no significant difference, SD not reported.
Percentage of patient without analgesic: no significant difference, SD not reported.
Trial discontinuation: active 31/50, control 31/49 (nausea and diarrhoea: active 6/50, control 7/49, progression: active 10/50, control 14/49, death: active 15/50, control 10/49)
No significant difference in median survival and survival rate at 6 month between active and control group.
No significant difference in number of sites of bone metastases at 0 and 6 month between active and control group.
Mean (SD) PSA at 0,1,3,6 months: active 335(563), 192(225), 213(280), 448(708), control 240(297), 201(263), 254(704), 214(229). P not significant between active and control group.

Notes

Kylmala 1997

Methods

Randomized double‐blind placebo‐controlled study
Quality score (1/2/0)
Primary outcome: pain
Other outcomes: use of analgesics, performance status, clinical response (i.e. better, same, worse by doctor), imaging response, PSA response, biochemical markers, side‐effects.
Pain assessment criteria: verbal ordinal scale 0‐4 by doctor and visual analogue scale by patient.
Definition of pain response: not defined.
Analgesic measurement: 3 point scale (0 none, 1 non‐narcotics < tid, 2 non‐narcotic > tid, 3 narcotic).
Pain assessment schedule: 0,1,3,6,12 months

Participants

Prostate cancer (failed hormonal therapy). Bone metastases required. No pain required.
Life expectancy 6 months or more.
Exclusion radiotherapy within 2 weeks.
Hormonal/ chemotherapy allowed.
Randomized 57 patients: active 28, placebo 29, analysed: active 28, control 27
Baseline pain (grade 0,1,2,3,4): active 6,13,5,3,0/28 control 2,12,12,1,0/29

Interventions

Active arm: clodronate (i.v. 300mg/day x 5days followed by oral 1.6g/day x 12 months)
Control arm: placebo
All received estramustine 280mg twice daily.

Outcomes

Pain: no significant different in any pain score. Proportion of patient with no pain at 1 month: active 10/28, control 6/29.
Analgesic: proportion of patient without analgesic at 1 month: active 13/28, control 8/29.
Performance status: proportion of patients asymptomatic at 1 month: active 4/28 control 1/29.
Clinical response: no significant difference.
Radiological response at 6 months: active 15/28 (5SD, 9PR, 1CR), control 14/29 (6SD, 7PR, 1CR)
Mean(SE) PSA at 0,1,3 months: active 161(52) 138(54) 226(70) control 214(104) 258(197) 331(204).
Adverse effects:
Nausea at 1 month: active 33% placebo 40%.
No renal failure

Notes

Saad 2002

Methods

Randomized double blind placebo‐controlled study.
3 arm study: 2 active arms, 1 placebo arm.
Quality score (2/2/1)
Primary outcome: skeletal‐related event (pathologic fracture, spinal cord compression, change in anti‐neoplastic therapy for bone pain, bone surgery or radiotherapy).
Secondary outcomes: time to first skeletal‐related event, skeletal morbidity rate, proportion of patients with skeletal‐related events, time to disease progression, bone lesion response, bone biochemical markers, quality of life parameters.
Pain assessment: Brief Pain Inventory pain score
Analgesic assessment: analgesic score 0‐4
Pain assessment schedule: 3,6,9,12,15 months

Participants

Prostate cancer (hormone refractory). Bone metastases required. Pain not required.
ECOG performance status of 0,1,2.
Cointervention: Antineoplastic therapy allowed.
Exclusion: bone pain requiring strong narcotic therapy, chemotherapy (except estramustine), RT within 3 months, previous bisphosphonate treatment, severe heart disease or hypertension, a serum creatinine of more than 3mg/dL or calcium less than 8mg/dL or greater than 11.6mg/dL.
Randomized 643 patients: active arm‐1 214, active arm‐2 221, control 208
Baseline pain, Mean BPI, active1 = 2 +/‐ 2, active2 = 2.5 +/‐ 2.1, control 2.1 +/‐ 2.0

Interventions

Active arm 1: zoledronic acid 4mg, 214 patients
Active arm 2: zoledronic acid 8mg, 221 patients
Control arm: placebo q3w x 20 cycles (15 months), 208 patients..
Ca supplement and vit D.

Outcomes

Pain score, mean change (95% confidence intervals) at 3 months: active1 ‐0.028 (0.22), active2 ‐0.04 (0.21), control 0.42 (0.22)
Analgesic score: No statistically significant difference (scores not included).
Patients having skeletal‐related events: active1 71/214, active2 85/221, control 92/208, pathologic fractures 28/214 33/221 46/208, vertebral fractures 8/214, 8/221, 17/208, non‐vertebral fractures 22/214, 22/221, 33/208, bone radiotherapy 49/214 53/221 61/208, bone surgery 5/214 6/221 7/208, spinal cord compression 9/214, 11/221, 14/208.
Time to first skeletal‐related event: active1 but not active2 is significantly longer than control.
Patients with radiological bone response: active1 56/214 (CR0,PR9,SD47), active2 52/221 (CR0,PR6,SD46), control 43/208 (CR0,PR8,SD35)
Patients with adverse events: bone pain (active1 108/214, active2 133/218, placebo 127/208), nausea (active1 77/214, active2 115/218, placebo 77/208), constipation, fatigue, anaemia (active1 57/214, active2 60/218, placebo 37/208), myalgia, vomiting (active1 46/214, active2 115/218 placebo 43/208), weakness, anorexia, fever, lower limb edema, dizziness, diarrhoea, weight increase.
ECOG performance scores, FACT‐GQoL and EURO‐QOL scores: No statistically significant difference.
Medican time to cancer progression = 84 days in all treatment groups.
PSA, percent change from baseline, no statistically significant difference.
Median survival (days) (active1, active2, placebo): 546, 407, 464
P = 0.091 (active1 vs placebo), P = 0.386 (active2 vs placebo).

Notes

Small 2003

Methods

Randomized double‐blind, placebo‐controlled trial.
Quality score (1/2/1)
Primary outcomes: pain and analgesic use
Pain assessment: Brief Pain Inventory (11 point numeric rating scale 0‐10) (least, average and worst pain since last visit)
Analgesic assessment: Oral morphine equivalent (OME)
Definition of pain and analgesic responses: mean change from baseline BPI and OME scores
Pain assessment schedule: 9 and 27 weeks. Study end at 27 weeks.
Secondary: skeletal related event, skeletal morbidity rate, mobility, bone imaging, PSA and other biochemical markers and disease progression (stage, clinical and pain).

Participants

Prostate cancer (hormone refractory). Bone metastases. Pain required.
Life expectancy 6 months or above. RT and radio‐isotope allowed.
Exclusion: leucopenia, thrombocytopenia, serum Cr 5mg/dL or above, corrected serum Ca 11mg/dL or above or 8.4mg/dL or below, magnesium 0.8mg/dL or below, or total bilirubin more than 2.5mg/dL. Patient with untreated brain metastases, prior bisphosphonate therapy, abnormal ECG, ascites, impending spinal cord compression or spinal orthosis, or skeletal event within 1 month. Change in chemotherapy or hormone therapy within 6 weeks. Receiving drugs that affected osteoclast activity.
Randomized 378 patients: active 182, control 196
Analyzed patients: active 169, control 181
Baseline pain, mean BPI, active 4.3 +/‐ 2.1, control 4.5 +/‐ 2.1

Interventions

Active arm: Pamidronate 90mg in 250ml D5 iv infusion over 2h q3w x 27 weeks.
Control arm: placebo

Outcomes

Mean Pain change (average BPI) at week 9: active ‐0.61 SE 0.17, n=147; placebo: ‐0.44 SE 0.16 n=154
OME score, week 9: active +16.6 SE 6.8, n=147; placebo: +10.2 SE 6.6 n=154
Decreasing or stable analgesic use at week 9: active 53/147, control 68/154; week 27: active 46/110, control 52/108.
Patients having SRE, week 9: active 20/169 control 20/181; week 27: active 42/169 control 46/181
Patients having RT for bone pain, week 9: active 11/169 control 10/181; week 27: active 25/169 control 29/181
Patients having RT to prevent fracture, week 9: active 6/169 control 3/181; week 27: active 8/169 control 7/181
Patients with nonvertebral fractures, week 9: active 6/169 control 6/181; week 27: active 14/169 control 12/181
Patients with vertebral fractures, week 9: active 5/169 control 2/181; week 27: active 11/169 control 10/181
Patients with spinal cord compression, week 9: active 3/169 control 0/181; week 27: active 5/169 control 3/181
Patients with surgery to bone, week 9: active 2/169 control 3/181; week 27: active 5/169 control 6/181
Patients with hypercalcaemia, week 9: active 0/169 control 1/181; week 27: active 1/169 control 2/181
Patients with adverse effects: bone pain (active 77/180, control 75/194); nausea (active 50/180, control 43/194); fatigue, anorexia, constipation, vomiting (active 31/180, control 31/194), anaemia (active 38/180, control 39/194), fever, weight decrease, asthenia, diarrhoea, dyspnoea, urinary tract infection, dizziness.
Mean PSA increase from baseline (data not shown)
All deaths: active 22 control 26 (primarily due to progressive disease)

Notes

Data pooled from two trials

Smith 1989

Methods

Randomized double‐blind placebo‐controlled study
3 active arms, 1 placebo arm
Quality score (1/1/0)
Outcomes: pain relief and analgesic requirement.
Pain assessment: numerical and linear analogue scales, rated by patient and investigator.
Definition of pain response: minor and major improvement, details not described.
Analgesic requirement: daily assessment, details not described.
Pain assessment schedule: daily pain diary by patient.

Participants

Prostate cancer (previous hormone therapy). Bone metastases required (documented by bone scan). Pain required.
Cointervention: hormonal therapy as clinically indicated. RT not allowed.
Exclusion: renal dysfunction
Randomized 57 patients: active1 14, active2 14, active3 15, control 14; evaluable: active1 13, active2 12, active3 14, control 12.

Interventions

Active 1 ‐ sodium etidronate 7.5mg/kg x 3 days then oral sodium etidronate 200mg bid
Active 2 ‐ sodium etidronate 7.5mg/kg x 3 days then placebo bid
Active 3 ‐ i.v. placebo then oral sodium etidronate 200mg bid
Control ‐ i.v. and oral placebo
Duration of treatment at least 1 month up to 6 month if responding.

Outcomes

Major pain improvement: active1 0/14, active2 2/14, active3 1/15, control 1/14
Minor pain improvement: active1 2/14, active2 2/14, active3 1/15, control 1/14
Decrease analgesic requirement: active 4/43, control 3/14.
Discontinued (2 due to nausea and vomiting): active1: 1/13, active2 2/12, active3 1/14, control 2/12
No serious adverse effects observed.
Mild stomach cramp and nausea: active 7/43, control 3/14.

Notes

Strang 1997

Methods

Randomized double‐blind placebo‐controlled study
Quality score (1/0/0)
Outcomes: pain intensity and analgesic consumption.
Pain assessment: mean pain, least pain, worst pain with 10cm VAS
Pain response definition: not specified
Analgesic consumption definition: not specified
Pain assessment schedule: Day 0,1,2,3,4,11,18,25,32.

Participants

Prostate cancer, hormone refractory, bone metastases.
Pain required, > 20mm VAS.
Conintervention: none described.
Life expectancy > 3m
Exclusion: renal dysfunction, bisphosphonate within previous 30 days, RT within previous 3 weeks.
Included 55 patients, randomized 52 patients (active 25, control 27), 46 patients valid for efficacy (meaning not defined, active 22, control 24).
Baseline pain, mean VAS (mm), active 39 +/‐ 19, control 53 +/‐ 21

Interventions

Active arm: Clodronate 300mg/day iv x 3 days then Clodroate 1600mg bd oral for 4 weeks
Control arm: isotonic saline for 3 days then placebo capsules

Outcomes

Baseline mean overall pain and mean pain during the best and worst period reported.
No significant changes in mean pain intensity and in pain intensity during best and worst period. Data not included.
Subgroup analysis (patients with baseline mean pain > 50mm VAS): also not significant pain reduction compared with control.
Analgesic consumption not reported.
Withdraws: not reported.

Notes

Study closed early due to lack of accrual.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Adami 1985

No control arm.

Carey 1988

No control arm.

Clarke 1991

No control arm.

Cresswell 1995

No control arm.

Fernandez‐Conde 1997

Randomized controlled study with histomorphometric outcomes. Pain not one of the outcomes.

Heidenreich 2001

Nonrandomized study

Heidenreich 2002

Nonrandomized study

Jagdev 2001

Randomised study comparing i.v. pamidronate with oral clodronate in a mixed tumour population. Not specific for prostate cancer.

Kylmala 1994

No control arm.

Magnusson 1998

Randomized controlled study with biochemical outcomes, clinical outcomes including pain were reported in another article by Strang 1997, one of the included studies.

Pelger 1998

No control arm.

Taube 1994

Randomized controlled study with histomorphometric outcomes. Pain not one of the outcomes.

Vorreuther 1992

No control arm.

Vorreuther 1993

No control arm.

Data and analyses

Open in table viewer
Comparison 1. Pain response

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients with pain response (intention‐to‐treat analysis) Show forest plot

4

416

Odds Ratio (M‐H, Fixed, 95% CI)

1.54 [0.97, 2.44]

Analysis 1.1

Comparison 1 Pain response, Outcome 1 Proportion of patients with pain response (intention‐to‐treat analysis).

Comparison 1 Pain response, Outcome 1 Proportion of patients with pain response (intention‐to‐treat analysis).

2 Proportion of patients with pain response (evaluable patients as defined by original studies) Show forest plot

4

374

Odds Ratio (M‐H, Fixed, 95% CI)

1.64 [1.02, 2.61]

Analysis 1.2

Comparison 1 Pain response, Outcome 2 Proportion of patients with pain response (evaluable patients as defined by original studies).

Comparison 1 Pain response, Outcome 2 Proportion of patients with pain response (evaluable patients as defined by original studies).

3 Mean pain change Show forest plot

2

723

Std. Mean Difference (IV, Fixed, 95% CI)

‐1.58 [‐1.75, ‐1.41]

Analysis 1.3

Comparison 1 Pain response, Outcome 3 Mean pain change.

Comparison 1 Pain response, Outcome 3 Mean pain change.

Open in table viewer
Comparison 2. Analgesic consumption

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients with decreased analgesic consumption Show forest plot

4

416

Odds Ratio (M‐H, Fixed, 95% CI)

1.27 [0.82, 1.98]

Analysis 2.1

Comparison 2 Analgesic consumption, Outcome 1 Proportion of patients with decreased analgesic consumption.

Comparison 2 Analgesic consumption, Outcome 1 Proportion of patients with decreased analgesic consumption.

Open in table viewer
Comparison 3. Skeletal events

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients having any skeletal events Show forest plot

3

1332

Odds Ratio (M‐H, Fixed, 95% CI)

0.79 [0.62, 1.00]

Analysis 3.1

Comparison 3 Skeletal events, Outcome 1 Proportion of patients having any skeletal events.

Comparison 3 Skeletal events, Outcome 1 Proportion of patients having any skeletal events.

2 Proportion of patients having any skeletal events (evaluable patients as defined by original studies) Show forest plot

3

1304

Odds Ratio (M‐H, Fixed, 95% CI)

0.79 [0.62, 1.00]

Analysis 3.2

Comparison 3 Skeletal events, Outcome 2 Proportion of patients having any skeletal events (evaluable patients as defined by original studies).

Comparison 3 Skeletal events, Outcome 2 Proportion of patients having any skeletal events (evaluable patients as defined by original studies).

3 Proportion of patients having any skeletal events (evaluable patients, selecting 8/4mg arm from Saad 2002) Show forest plot

3

1090

Odds Ratio (M‐H, Fixed, 95% CI)

0.84 [0.65, 1.08]

Analysis 3.3

Comparison 3 Skeletal events, Outcome 3 Proportion of patients having any skeletal events (evaluable patients, selecting 8/4mg arm from Saad 2002).

Comparison 3 Skeletal events, Outcome 3 Proportion of patients having any skeletal events (evaluable patients, selecting 8/4mg arm from Saad 2002).

4 Proportion of patients having any skeletal events (evaluable patients, selecting 4mg arm from Saad 2002) Show forest plot

3

1083

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.59, 0.98]

Analysis 3.4

Comparison 3 Skeletal events, Outcome 4 Proportion of patients having any skeletal events (evaluable patients, selecting 4mg arm from Saad 2002).

Comparison 3 Skeletal events, Outcome 4 Proportion of patients having any skeletal events (evaluable patients, selecting 4mg arm from Saad 2002).

5 Proportion of patients having pathologic fractures Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.75 [0.53, 1.06]

Analysis 3.5

Comparison 3 Skeletal events, Outcome 5 Proportion of patients having pathologic fractures.

Comparison 3 Skeletal events, Outcome 5 Proportion of patients having pathologic fractures.

6 Proportion of patients having vertebral fractures Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.65 [0.38, 1.13]

Analysis 3.6

Comparison 3 Skeletal events, Outcome 6 Proportion of patients having vertebral fractures.

Comparison 3 Skeletal events, Outcome 6 Proportion of patients having vertebral fractures.

7 Proportion of patients having non‐vertebral fractures Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.74 [0.49, 1.12]

Analysis 3.7

Comparison 3 Skeletal events, Outcome 7 Proportion of patients having non‐vertebral fractures.

Comparison 3 Skeletal events, Outcome 7 Proportion of patients having non‐vertebral fractures.

8 Proportion of patients having spinal cord compression Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.82 [0.44, 1.55]

Analysis 3.8

Comparison 3 Skeletal events, Outcome 8 Proportion of patients having spinal cord compression.

Comparison 3 Skeletal events, Outcome 8 Proportion of patients having spinal cord compression.

9 Proportion of patients received bone radiotherapy Show forest plot

3

1248

Odds Ratio (M‐H, Fixed, 95% CI)

0.83 [0.62, 1.11]

Analysis 3.9

Comparison 3 Skeletal events, Outcome 9 Proportion of patients received bone radiotherapy.

Comparison 3 Skeletal events, Outcome 9 Proportion of patients received bone radiotherapy.

10 Proportion of patients received bone surgery Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.38, 1.70]

Analysis 3.10

Comparison 3 Skeletal events, Outcome 10 Proportion of patients received bone surgery.

Comparison 3 Skeletal events, Outcome 10 Proportion of patients received bone surgery.

Open in table viewer
Comparison 4. Treatment response

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients died of prostate cancer Show forest plot

4

991

Odds Ratio (M‐H, Fixed, 95% CI)

0.82 [0.61, 1.11]

Analysis 4.1

Comparison 4 Treatment response, Outcome 1 Proportion of patients died of prostate cancer.

Comparison 4 Treatment response, Outcome 1 Proportion of patients died of prostate cancer.

2 Proportion of patients with disease progression Show forest plot

2

538

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.53, 1.08]

Analysis 4.2

Comparison 4 Treatment response, Outcome 2 Proportion of patients with disease progression.

Comparison 4 Treatment response, Outcome 2 Proportion of patients with disease progression.

3 Proportion of patients with radiological response Show forest plot

2

700

Odds Ratio (M‐H, Fixed, 95% CI)

1.26 [0.87, 1.83]

Analysis 4.3

Comparison 4 Treatment response, Outcome 3 Proportion of patients with radiological response.

Comparison 4 Treatment response, Outcome 3 Proportion of patients with radiological response.

Open in table viewer
Comparison 5. Performance status

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients with improvement in performance status Show forest plot

2

368

Odds Ratio (M‐H, Fixed, 95% CI)

1.53 [0.81, 2.91]

Analysis 5.1

Comparison 5 Performance status, Outcome 1 Proportion of patients with improvement in performance status.

Comparison 5 Performance status, Outcome 1 Proportion of patients with improvement in performance status.

Open in table viewer
Comparison 6. Adverse events

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients having nausea Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

1.35 [1.02, 1.77]

Analysis 6.1

Comparison 6 Adverse events, Outcome 1 Proportion of patients having nausea.

Comparison 6 Adverse events, Outcome 1 Proportion of patients having nausea.

2 Proportion of patients having vomiting Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

1.22 [0.89, 1.69]

Analysis 6.2

Comparison 6 Adverse events, Outcome 2 Proportion of patients having vomiting.

Comparison 6 Adverse events, Outcome 2 Proportion of patients having vomiting.

3 Proportion of patients having anaemia Show forest plot

3

1037

Odds Ratio (M‐H, Fixed, 95% CI)

1.04 [0.76, 1.41]

Analysis 6.3

Comparison 6 Adverse events, Outcome 3 Proportion of patients having anaemia.

Comparison 6 Adverse events, Outcome 3 Proportion of patients having anaemia.

4 Proportion of patients having bone pain Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.93 [0.72, 1.21]

Analysis 6.4

Comparison 6 Adverse events, Outcome 4 Proportion of patients having bone pain.

Comparison 6 Adverse events, Outcome 4 Proportion of patients having bone pain.

Comparison 1 Pain response, Outcome 1 Proportion of patients with pain response (intention‐to‐treat analysis).
Figuras y tablas -
Analysis 1.1

Comparison 1 Pain response, Outcome 1 Proportion of patients with pain response (intention‐to‐treat analysis).

Comparison 1 Pain response, Outcome 2 Proportion of patients with pain response (evaluable patients as defined by original studies).
Figuras y tablas -
Analysis 1.2

Comparison 1 Pain response, Outcome 2 Proportion of patients with pain response (evaluable patients as defined by original studies).

Comparison 1 Pain response, Outcome 3 Mean pain change.
Figuras y tablas -
Analysis 1.3

Comparison 1 Pain response, Outcome 3 Mean pain change.

Comparison 2 Analgesic consumption, Outcome 1 Proportion of patients with decreased analgesic consumption.
Figuras y tablas -
Analysis 2.1

Comparison 2 Analgesic consumption, Outcome 1 Proportion of patients with decreased analgesic consumption.

Comparison 3 Skeletal events, Outcome 1 Proportion of patients having any skeletal events.
Figuras y tablas -
Analysis 3.1

Comparison 3 Skeletal events, Outcome 1 Proportion of patients having any skeletal events.

Comparison 3 Skeletal events, Outcome 2 Proportion of patients having any skeletal events (evaluable patients as defined by original studies).
Figuras y tablas -
Analysis 3.2

Comparison 3 Skeletal events, Outcome 2 Proportion of patients having any skeletal events (evaluable patients as defined by original studies).

Comparison 3 Skeletal events, Outcome 3 Proportion of patients having any skeletal events (evaluable patients, selecting 8/4mg arm from Saad 2002).
Figuras y tablas -
Analysis 3.3

Comparison 3 Skeletal events, Outcome 3 Proportion of patients having any skeletal events (evaluable patients, selecting 8/4mg arm from Saad 2002).

Comparison 3 Skeletal events, Outcome 4 Proportion of patients having any skeletal events (evaluable patients, selecting 4mg arm from Saad 2002).
Figuras y tablas -
Analysis 3.4

Comparison 3 Skeletal events, Outcome 4 Proportion of patients having any skeletal events (evaluable patients, selecting 4mg arm from Saad 2002).

Comparison 3 Skeletal events, Outcome 5 Proportion of patients having pathologic fractures.
Figuras y tablas -
Analysis 3.5

Comparison 3 Skeletal events, Outcome 5 Proportion of patients having pathologic fractures.

Comparison 3 Skeletal events, Outcome 6 Proportion of patients having vertebral fractures.
Figuras y tablas -
Analysis 3.6

Comparison 3 Skeletal events, Outcome 6 Proportion of patients having vertebral fractures.

Comparison 3 Skeletal events, Outcome 7 Proportion of patients having non‐vertebral fractures.
Figuras y tablas -
Analysis 3.7

Comparison 3 Skeletal events, Outcome 7 Proportion of patients having non‐vertebral fractures.

Comparison 3 Skeletal events, Outcome 8 Proportion of patients having spinal cord compression.
Figuras y tablas -
Analysis 3.8

Comparison 3 Skeletal events, Outcome 8 Proportion of patients having spinal cord compression.

Comparison 3 Skeletal events, Outcome 9 Proportion of patients received bone radiotherapy.
Figuras y tablas -
Analysis 3.9

Comparison 3 Skeletal events, Outcome 9 Proportion of patients received bone radiotherapy.

Comparison 3 Skeletal events, Outcome 10 Proportion of patients received bone surgery.
Figuras y tablas -
Analysis 3.10

Comparison 3 Skeletal events, Outcome 10 Proportion of patients received bone surgery.

Comparison 4 Treatment response, Outcome 1 Proportion of patients died of prostate cancer.
Figuras y tablas -
Analysis 4.1

Comparison 4 Treatment response, Outcome 1 Proportion of patients died of prostate cancer.

Comparison 4 Treatment response, Outcome 2 Proportion of patients with disease progression.
Figuras y tablas -
Analysis 4.2

Comparison 4 Treatment response, Outcome 2 Proportion of patients with disease progression.

Comparison 4 Treatment response, Outcome 3 Proportion of patients with radiological response.
Figuras y tablas -
Analysis 4.3

Comparison 4 Treatment response, Outcome 3 Proportion of patients with radiological response.

Comparison 5 Performance status, Outcome 1 Proportion of patients with improvement in performance status.
Figuras y tablas -
Analysis 5.1

Comparison 5 Performance status, Outcome 1 Proportion of patients with improvement in performance status.

Comparison 6 Adverse events, Outcome 1 Proportion of patients having nausea.
Figuras y tablas -
Analysis 6.1

Comparison 6 Adverse events, Outcome 1 Proportion of patients having nausea.

Comparison 6 Adverse events, Outcome 2 Proportion of patients having vomiting.
Figuras y tablas -
Analysis 6.2

Comparison 6 Adverse events, Outcome 2 Proportion of patients having vomiting.

Comparison 6 Adverse events, Outcome 3 Proportion of patients having anaemia.
Figuras y tablas -
Analysis 6.3

Comparison 6 Adverse events, Outcome 3 Proportion of patients having anaemia.

Comparison 6 Adverse events, Outcome 4 Proportion of patients having bone pain.
Figuras y tablas -
Analysis 6.4

Comparison 6 Adverse events, Outcome 4 Proportion of patients having bone pain.

Comparison 1. Pain response

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients with pain response (intention‐to‐treat analysis) Show forest plot

4

416

Odds Ratio (M‐H, Fixed, 95% CI)

1.54 [0.97, 2.44]

2 Proportion of patients with pain response (evaluable patients as defined by original studies) Show forest plot

4

374

Odds Ratio (M‐H, Fixed, 95% CI)

1.64 [1.02, 2.61]

3 Mean pain change Show forest plot

2

723

Std. Mean Difference (IV, Fixed, 95% CI)

‐1.58 [‐1.75, ‐1.41]

Figuras y tablas -
Comparison 1. Pain response
Comparison 2. Analgesic consumption

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients with decreased analgesic consumption Show forest plot

4

416

Odds Ratio (M‐H, Fixed, 95% CI)

1.27 [0.82, 1.98]

Figuras y tablas -
Comparison 2. Analgesic consumption
Comparison 3. Skeletal events

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients having any skeletal events Show forest plot

3

1332

Odds Ratio (M‐H, Fixed, 95% CI)

0.79 [0.62, 1.00]

2 Proportion of patients having any skeletal events (evaluable patients as defined by original studies) Show forest plot

3

1304

Odds Ratio (M‐H, Fixed, 95% CI)

0.79 [0.62, 1.00]

3 Proportion of patients having any skeletal events (evaluable patients, selecting 8/4mg arm from Saad 2002) Show forest plot

3

1090

Odds Ratio (M‐H, Fixed, 95% CI)

0.84 [0.65, 1.08]

4 Proportion of patients having any skeletal events (evaluable patients, selecting 4mg arm from Saad 2002) Show forest plot

3

1083

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.59, 0.98]

5 Proportion of patients having pathologic fractures Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.75 [0.53, 1.06]

6 Proportion of patients having vertebral fractures Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.65 [0.38, 1.13]

7 Proportion of patients having non‐vertebral fractures Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.74 [0.49, 1.12]

8 Proportion of patients having spinal cord compression Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.82 [0.44, 1.55]

9 Proportion of patients received bone radiotherapy Show forest plot

3

1248

Odds Ratio (M‐H, Fixed, 95% CI)

0.83 [0.62, 1.11]

10 Proportion of patients received bone surgery Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.80 [0.38, 1.70]

Figuras y tablas -
Comparison 3. Skeletal events
Comparison 4. Treatment response

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients died of prostate cancer Show forest plot

4

991

Odds Ratio (M‐H, Fixed, 95% CI)

0.82 [0.61, 1.11]

2 Proportion of patients with disease progression Show forest plot

2

538

Odds Ratio (M‐H, Fixed, 95% CI)

0.76 [0.53, 1.08]

3 Proportion of patients with radiological response Show forest plot

2

700

Odds Ratio (M‐H, Fixed, 95% CI)

1.26 [0.87, 1.83]

Figuras y tablas -
Comparison 4. Treatment response
Comparison 5. Performance status

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients with improvement in performance status Show forest plot

2

368

Odds Ratio (M‐H, Fixed, 95% CI)

1.53 [0.81, 2.91]

Figuras y tablas -
Comparison 5. Performance status
Comparison 6. Adverse events

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Proportion of patients having nausea Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

1.35 [1.02, 1.77]

2 Proportion of patients having vomiting Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

1.22 [0.89, 1.69]

3 Proportion of patients having anaemia Show forest plot

3

1037

Odds Ratio (M‐H, Fixed, 95% CI)

1.04 [0.76, 1.41]

4 Proportion of patients having bone pain Show forest plot

2

1021

Odds Ratio (M‐H, Fixed, 95% CI)

0.93 [0.72, 1.21]

Figuras y tablas -
Comparison 6. Adverse events