Scolaris Content Display Scolaris Content Display

Effect of testing for cancer on cancer‐ and venous thromboembolism (VTE)‐related mortality and morbidity in people with unprovoked VTE

This is not the most recent version

Collapse all Expand all

References

References to studies included in this review

Carrier 2015 {published data only}

Carrier M, Lazo‐Langner A, Shivakumar S, Tagalakis V, Zarychanski R, Solymoss S, et al. Screening for occult cancer in unprovoked venous thromboembolism. New England Journal of Medicine 2015;373(8):697‐704. CENTRAL

Piccioli 2004b {published data only}

Otten HM, Prins MH. A number needed to screen and cost‐effectiveness analysis of the SOMIT‐data. Haemostasis 2001;31 Suppl 1:40‐2. CENTRAL
Piccioli A, Lensing AWA, Prins MH, Falanga A, Scannapieco GL, Ieran M, et al. Extensive screening for occult malignant disease in idiopathic venous thromboembolism: a prospective randomized clinical trial. Journal of Thrombosis and Haemostasis 2004;2:884‐9. CENTRAL

Prandoni 2016 {published data only}

Piccioli A. The value of CT scanning for detection of occult cancer in patients with idiopathic VTE. Cancer, thrombosis and low‐molecular‐weight heparins (dare.uva.nl/record/1/465654 (accessed June 2016)). Amsterdam (the Netherlands): University of Amsterdam, 2015. CENTRAL
Piccioli A, Bernardi E, Dalla Valle F, Visona A, Tropeano F, Bova C, et al. The value of CT‐scanning for detection of occult cancer in patients with unprovoked venous thromboembolism. The D'Acquapendente study. Journal of Thrombosis and Haemostasis 2013;11 (Suppl 2):50‐1. CENTRAL
Piccioli A, Bernardi E, Dalla Valle F, Visonà A, Tropeano PF, Bova C, et al. The value of thoraco‐abdominal CT scanning for the detection of occult cancer in patients with unprovoked venous thromboembolism. A randomized study. Thrombosis Research 2012;129 Suppl 1:S155‐94. CENTRAL
Prandoni P, Bernardi E, Valle FD, Visona A, Tropeano PF, Bova C, et al. Extensive computed tomography versus limited screening for detection of occult cancer in unprovoked venous thromboembolism: a multicenter, controlled, randomized clinical trial. Seminars in Thrombosis and Hemostasis 2016;42(8):884‐90. CENTRAL

Robin 2016 {published data only}

Robin P, Le Roux PY, Planquette B, Accassat S, Roy PM, Couturaud F, et al. Limited screening with versus without (18)F‐fluorodeoxyglucose PET/CT for occult malignancy in unprovoked venous thromboembolism: an open‐label randomised controlled trial. Lancet Oncology 2016;17(2):193‐9. CENTRAL

Additional references

Atkins 2004

Atkins D, Best D, Briss PA, Eccles M, Falck‐Ytter Y, Flottorp S, et al. GRADE Working Group. Grading quality of evidence and strength of recommendations. BMJ 2004;328(7454):1490‐4.

Baron 1998

Baron JA, Gridley G, Weiderpass E, Nyren O, Linet M. Venous thromboembolism and cancer. Lancet 1998;351(9109):1077‐80.

Bick 1978

Bick RL. Alterations of hemostasis associated with malignancy: etiology, pathophysiology, diagnosis and management. Seminars in Thrombosis and Hemostasis 1978;5(1):1‐26.

Blann 2006

Blann AD, Lip GYH. Venous thromboembolism. BMJ 2006;332(7535):215‐9.

Carrier 2014

Carrier M, Cameron C, Delluc A, Castellucci L, Khorana AA, Lee AY. Efficacy and safety of anticoagulant therapy for the treatment of acute cancer‐associated thrombosis: a systematic review and meta‐analysis. Thrombosis Research 2014;134(6):1214‐9.

Deeks 2011

Deeks JJ, Higgins JPT, Altman DG. Chapter 9: Analysing data and undertaking meta‐analyses. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC. Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Kakkar 2003

Kakkar AK, Levine M, Pinedo HM, Wolff R, Wong J. Venous thrombosis in cancer patients: insights from the FRONTLINE survey. Oncologist 2003;8(4):381‐8.

Lee 2003a

Lee AY, Levine MN. Venous thromboembolism and cancer: risks and outcomes. Circulation 2003;107(23 Suppl 1):I17‐21.

Lee 2003b

Lee AYY, Levine MN, Baker RI, Bowden C, Kakkar AK, Prins M, et al. Low‐molecular‐weight heparin versus a coumarin for the prevention of recurrent venous thromboembolism in patients with cancer. New England Journal of Medicine 2003;349(2):146‐53.

Levitan 1999

Levitan N, Dowlati A, Remick SC, Tahsildar HI, Sivinski LD, Beyth R, et al. Rates of initial and recurrent thromboembolic disease among patients with malignancy versus those without malignancy. Risk analysis using Medicare claims data. Medicine (Baltimore) 1999;78(5):285‐91.

NICE 2012

National Institute for Health and Care Excellence. Venous thromboembolic diseases: the management of venous thromboembolic diseases and the role of thrombophilia testing. NICE Clinical Guideline 144. www.nice.org.uk/guidance/cg144 (accessed 2 August 2017).

Piccioli 2004a

Piccioli A, Lensing AW, Prins MH, Falanga A, Scannapieco GL, Ieran M, et al. SOMIT Investigators Group. Extensive screening for occult malignant disease in idiopathic venous thromboembolism: a prospective randomized clinical trial. Journal of Thrombosis and Haemostasis 2004;2(6):884‐9.

Prandoni 1997

Prandoni P, Piccioli A. Venous thromboembolism and cancer: a two‐way clinical association. Frontiers in Bioscience 1997;2:e12‐21.

RevMan 2014 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Sorensen 2000

Sorensen HT, Mellemkjaer L, Olsen JH, Baron JA. Prognosis of cancers associated with venous thromboembolism. New England Journal of Medicine 2000;343:1846‐50.

Sterne 2011

Sterne JAC, Egger M, Moher D. Chapter 10: Addressing reporting biases. In: Higgins JPT, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions. Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Vedovati 2015

Vedovati MC, Germini F, Agnelli G, Becattini C. Direct oral anticoagulants in patients with VTE and cancer: a systematic review and meta‐analysis. Chest 2015;147(2):475‐83.

White 2005

White RH, Chew HK, Zhou H, Parikh‐Patel A, Harris D, Harvey D, et al. Incidence of venous thromboembolism in the year before the diagnosis of cancer in 528,693 adults. Archives of Internal Medicine 2005;165(15):1782‐7.

References to other published versions of this review

Robertson 2013

Robertson L, Agarwal R, Yeoh SE. Effect of testing for cancer on cancer‐ and venous thromboembolism (VTE)‐related mortality and morbidity in patients with unprovoked VTE. Cochrane Database of Systematic Reviews 2013, Issue 11. [DOI: 10.1002/14651858.CD010837]

Robertson 2015

Robertson L, Yeoh SE, Stansby G, Agarwal R. Effect of testing for cancer on cancer‐ and venous thromboembolism (VTE)‐related mortality and morbidity in patients with unprovoked VTE. Cochrane Database of Systematic Reviews 2015, Issue 3. [DOI: 10.1002/14651858.CD010837.pub2]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Carrier 2015

Methods

Study design: multicentre, open‐label, randomised controlled trial.

Source of funding: Heart and Stroke Foundation of Canada.

Participants

Country: Canada.

Setting: hospital.

Number of centres: 9.

Number of participants: 854.

Age (mean (SD)): screening + CT group: 53.4 (14.2) years; screening only group: 53.7 (13.8).

Sex: screening + CT group: 299 M/124 F; screening only group: 277 M/154 F.

Inclusion criteria: people with new diagnosis of first unprovoked VTE (proximal lower‐limb deep vein thrombosis, pulmonary embolism, or both). Unprovoked VTE defined as VTE in absence of known overt active cancer, current pregnancy, thrombophilia (hereditary or acquired), previous unprovoked VTE or a temporary predisposing factor in the previous 3 months, including paralysis, paresis or plaster immobilisation of the legs, confinement to bed for ≥ 3 days or major surgery.

Exclusion criteria: aged < 18 years, refusal or inability to provide informed consent, allergy to contrast media, creatinine clearance < 60 mL per minute, claustrophobia or agoraphobia, weight > 130 kg, ulcerative colitis or glaucoma.

Interventions

Screening procedure: complete history and physical examination, measurement of complete blood counts and serum electrolyte and creatinine levels, liver‐function testing and chest radiography. Sex‐specific screening conducted if it had not been performed in previous year. Breast examination, mammography, or both performed in women > 50 years of age and Pap testing and a pelvic examination performed in women 18‐70 years of age who had never been sexually active. Prostate examination, PSA test, or both performed in men aged > 40 years. Also comprehensive CT of abdomen and pelvis (virtual colonoscopy and gastroscopy, biphasic enhanced CT of liver, parenchymal pancreatography, and uniphasic enhanced CT of distended bladder).

Control: complete history and physical examination, measurement of complete blood counts and serum electrolyte and creatinine levels, liver‐function testing and chest radiography. Sex‐specific screening conducted if it had not been performed in previous year. Breast examination, mammography, or both performed in women > 50 years of age and Pap testing and a pelvic examination performed in women 18‐70 years of age who had ever been sexually active. Prostate examination, PSA test, or both performed in men aged > 40 years.

Duration: 1 year follow‐up.

Outcomes

Primary outcomes: newly diagnosed cancer during the follow‐up period in people who had a negative screening result for occult cancer.

Secondary outcomes: total number of occult cancers diagnosed and total number of early cancers (T1‐2, N0, M0 according to the World Health Organization TNM classification system) diagnosed by occult‐cancer screening and during subsequent 1‐year follow‐up, 1‐year cancer‐related mortality, 1‐year overall mortality, time to cancer diagnosis and incidence of recurrent VTE.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The trial statistician generated the randomisation list using random‐number tables."

Allocation concealment (selection bias)

Low risk

Quote: "A central Web‐based randomisation system ensured assignment concealment."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Comment: blinding of participants and study personnel not done but review authors judged that outcome and outcome measurement not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "A central adjudication committee whose members were unaware of the study‐group assignments reviewed all suspected outcome events."

Comment: outcome assessors blinded to study allocation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: all losses to follow‐up accounted for.

Selective reporting (reporting bias)

Low risk

Comment: primary and secondary outcomes clearly prespecified and reported.

Other bias

Low risk

Comment: study appeared free from other sources of bias.

Piccioli 2004b

Methods

Study design: randomised multicentre clinical trial.

Source of funding: Associazone Italiana per le Ricerca sul Cancro.

Participants

Country: Italy.

Setting: hospital.

Number of centres: not stated.

Number of participants: 201.

Age (mean (SD)): screening group: 66.2 (13.1) years; no screening group: 66.6 (13.1) years.

Sex: screening group: 54 M/45 F; no screening group: 46 M/56 F.

Inclusion criteria: apparently cancer‐free people with a documented unprovoked first episode of symptomatic deep vein thrombosis of the lower extremity or pulmonary embolism.

Exclusion criteria: recognised risk factor for VTE (malignant disease, trauma of the leg, surgical procedures or immobilisation within 6 months, confirmed spontaneous VTE in a first‐degree relative, deficiency of antithrombin, protein C or S, presence of circulating lupus anticoagulant, oestrogen use, pregnancy or childbirth), previously documented VTE, malignant disease identified at routine physical examination, history taking, laboratory assessment or chest X‐ray at referral, unable to attend follow‐up due to geographic inaccessibility and aged < 25 years.

Interventions

Screening procedure: combination of ultrasound and CT scan of abdomen and pelvis, gastroscopy or double‐contrast barium swallow, flexible sigmoidoscopy or rectoscopy followed by barium enema or colonoscopy, haemoccult, sputum cytology and tumour markers including carcinoembryonic antigen, α‐fetoprotein and CA125. In addition, women had gynaecological examination, Pap smear and mammography. Men had a transabdominal ultrasound of prostate and total PSA test.

Control: tests at physician's discretion.

Duration: 2‐year follow‐up. At these visits, special attention paid to recent medical history. To avoid diagnostic suspicion bias, medical history concerning general health, hospital admission, and occurrence of signs and symptoms of cancer obtained on a standardised form by a physician unaware of allocation of participant. If malignant disease had become apparent during follow‐up, information from the attending specialist sought after consent of participant.

Outcomes

Primary outcomes: cancer‐related mortality defined as death due to a malignant disease itself, or death due to complications of diagnostic or surgical procedures performed to diagnose or treat cancer.

Secondary outcomes: cluster of cancer‐related mortality and presence of objectively documented residual malignancy or recurrent malignancy at 24 months and sensitivity of the diagnostic work‐up for occult malignancy.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "According to the Zelen design, patients randomised to..."

Allocation concealment (selection bias)

Low risk

Quote: "Randomisation was performed centrally."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Patients randomised to extensive screening were informed about the study. As patients allocated to the control group were not informed about the study, patients and their physicians were not discouraged to search for malignant disease."

Comment: blinding of participants in extensive screening group and study personnel not done but review authors judged that outcome and outcome measurement not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote: "To avoid diagnostic suspicion bias, the medical history concerning general health, hospital admission and occurrence of signs and symptoms of cancer were obtained on a standardised form by a physician unaware of allocation of the patient."

Comment: outcome assessors blinded to study allocation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: all participants completed the 2‐year follow‐up. No missing data.

Selective reporting (reporting bias)

Low risk

Comment: primary and secondary outcomes clearly prespecified and reported.

Other bias

High risk

Comment: study terminated early after inclusion of only 201 participants after 5 years for several reasons. First, only 5 of the more than 40 potential participating centres could contribute participants to the study. Second, some medical ethics committees rejected the protocol because of the absence of screening for occult cancer in the control group, other centres could not start because the proposed extensive screening was judged to be unethical. Finally, identification of cancer at an apparent early stage in the extensive screening group led to an increasing tendency among physicians in participating hospitals to initiate screening for cancer in control participants.

Prandoni 2016

Methods

Study design: multicentre, randomised controlled trial.

Participants

Country: Italy.

Setting: hospital.

Number of centres: 5.

Number of participants: 195

Age (mean (SD)): extensive screening group: 69.3 (14) years; control group: 69.0 (14) years.

Sex: extensive screening group: 54 M/44 F; control group: 47 M/50 F.

Inclusion criteria: people with an objectively diagnosed, first episode of unprovoked VTE, in whom a routine initial screening for cancer was normal.

Exclusion criteria: history of previous documented episodes of VTE, aged < 18 years, pregnant, unable to attend follow‐up visits because of geographic inaccessibility, had known allergy to contrast medium or had a CT scan of torso for any reasons within 6 months from presentation.

Interventions

Screening procedure: extensive screening with mandatory CT scan of thorax, abdomen and pelvis together with haemoccult test or any test at physician's discretion according to good clinical practice.

Control: personalised strategy consisting of additional testing based on physicians' judgements and participants' preferences, including a 'no‐further testing' option.

Duration: 3, 6, 12 and 24 months' follow‐up in which participants were asked about general health, history of recent hospital admissions and occurrence of signs and symptoms suggestive of cancer. Cancer outcomes that presented during follow‐up were detected based on clinical features that would prompt diagnostic imaging or cancers that were occasionally detected by screening that was independent of the diagnosis of VTE.

Outcomes

Primary outcomes: cancer‐related mortality (defined as death due to malignancy or death due to the complications of the diagnostic or surgical procedures performed to diagnose or treat cancer) and incidence of newly discovered cancer.

Secondary outcomes: cancer stage, using the TNM classification, at which tumours were diagnosed in the 2 study groups; and incidence of cancer‐related mortality in the 2 randomisation groups.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Concealed allocation was ensured by employing serially numbered, opaque, sealed envelopes. Each participating centre was initially assigned a lot of 20 envelopes, while subsequent allocations were in lots of 10, as needed."

Allocation concealment (selection bias)

Low risk

Quote: "Concealed allocation was ensured by employing serially numbered, opaque, sealed envelopes. Each participating centre was initially assigned a lot of 20 envelopes, while subsequent allocations were in lots of 10, as needed."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Comment: blinding of participants in extensive screening group and study personnel not done but review authors judged that outcome and outcome measurement were not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Comment: investigators performing the follow‐up visits blinded to participants' randomisation groups'.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: all losses to follow‐up accounted for.

Selective reporting (reporting bias)

Low risk

Comment: primary and secondary outcomes clearly prespecified and reported.

Other bias

High risk

Comment: interim analysis scheduled after inclusion of approximately half of planned sample size. Based on results of this analysis, study promoters decided to stop study enrolment because of low recruitment rate and of failure to show an appreciable advantage of CT‐based strategy over control strategy for detection of occult cancers.

Robin 2016

Methods

Study design: open‐label, multicentre, randomised study.

Source of funding: Programme Hospitalier de Recherche Clinique (French Department of Health).

Participants

Country: France.

Setting: hospital.

Number of centres: 4.

Number of participants: 394.

Age (mean (range)): screening group: 64 (48‐77) years; limited screening group: 62 (50‐75) years.

Sex: screening group: 105 M/92 F; limited screening group: 102 M/95 F.

Inclusion criteria: aged ≥ 18 years, diagnosed with unprovoked VTE. VTE defined as objectively confirmed proximal deep vein thrombosis or pulmonary embolism. Unprovoked VTE defined as VTE not provoked by major inherited or acquired risk factor including surgery, trauma or fracture during 3 months before VTE event, known antiphospholipid antibody syndrome or known deficiency in antithrombin, protein C or protein S.

Exclusion criteria: ongoing pregnancy, active malignant disease (defined as known malignant disease which was active or treated during previous 5 years), not insured under French National Social Security programme, hypersensitivity to 18F‐FDG or any of the excipients according to summary of product characteristics in France, or unable or unwilling to give consent.

Interventions

Screening procedure: screening strategy consisting of limited strategy + 18F‐FDG PET/CT scan of chest, abdomen and pelvis.

Control: limited screening strategy (physical examination, usual laboratory tests and basic radiographs).

Duration: 2 years.

Outcomes

Primary outcomes: proportion of people with a cancer diagnosis in each group after the initial screening assessment.

Secondary outcomes: subsequent cancer diagnosis in people with negative initial screening, proportion of early‐stage versus advanced‐stage tumours at initial screening and during follow‐up, overall mortality and cancer‐related mortality during follow‐up.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The randomisation list was created centrally using computer‐generated block sizes of six, stratified by centre, and concealed from investigators. We used a secure, dedicated, central web‐based randomisation system (Clinsight)."

Allocation concealment (selection bias)

Low risk

Quote: "The randomisation list was created centrally using computer‐generated block sizes of six, stratified by centre, and concealed from investigators. We used a secure, dedicated, central web‐based randomisation system (Clinsight). A unique study participant number and study group allocation was given after patients’ basic information and eligibility criteria were entered by the study personnel."

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Participants and physicians giving the intervention, assessing outcomes, and analysing the data were not masked to study group assignment."

Comment: blinding of participants and study personnel not done but review authors judged that outcome and outcome measurement not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote: "Physicians giving the intervention, assessing outcomes, and analysing the data were not masked to study group assignment."

Comment: outcome assessors not blinded to outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Comment: all losses to follow‐up accounted for.

Selective reporting (reporting bias)

Low risk

Comment: primary and secondary outcomes clearly prespecified and reported.

Other bias

Low risk

Comment: study appeared free from other sources of bias.

CT: computed tomography; F: female; FDG: fluorodeoxyglucose; M: male; Pap: Papanicolaou; PET: positron emission tomography; PSA: prostate‐specific antigen; SD: standard deviation; TNM: tumour‐node‐metastasis; VTE: venous thromboembolism.

Data and analyses

Open in table viewer
Comparison 1. Extensive tests versus tests at the physician's discretion

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Cancer‐related mortality Show forest plot

2

396

Odds Ratio (M‐H, Random, 95% CI)

0.49 [0.15, 1.67]

Analysis 1.1

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 1 Cancer‐related mortality.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 1 Cancer‐related mortality.

2 Characteristics of diagnosed cancer: type of cancer Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 2 Characteristics of diagnosed cancer: type of cancer.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 2 Characteristics of diagnosed cancer: type of cancer.

2.1 Lung

1

201

Odds Ratio (M‐H, Random, 95% CI)

2.08 [0.19, 23.34]

2.2 Bladder

1

201

Odds Ratio (M‐H, Random, 95% CI)

2.08 [0.19, 23.34]

2.3 Stomach

1

201

Odds Ratio (M‐H, Random, 95% CI)

1.03 [0.06, 16.71]

2.4 Kidney

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.5 Adrenal gland

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.6 Liver

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.7 Uterus

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.8 Breast

1

201

Odds Ratio (M‐H, Random, 95% CI)

1.03 [0.06, 16.71]

2.9 Ovary

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.10 Colon

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.51 [0.05, 5.72]

2.11 Prostate

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.51 [0.05, 5.72]

2.12 Pancreas

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.20 [0.01, 4.26]

3 Characteristics of diagnosed cancer: stage of cancer Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 3 Characteristics of diagnosed cancer: stage of cancer.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 3 Characteristics of diagnosed cancer: stage of cancer.

3.1 T1 or T2 (N0 M0)

1

201

Odds Ratio (M‐H, Random, 95% CI)

5.00 [1.05, 23.76]

3.2 T3

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.25 [0.03, 2.28]

4 Frequency of underlying cancer diagnosis Show forest plot

2

396

Odds Ratio (M‐H, Random, 95% CI)

1.32 [0.59, 2.93]

Analysis 1.4

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 4 Frequency of underlying cancer diagnosis.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 4 Frequency of underlying cancer diagnosis.

Open in table viewer
Comparison 2. Standard testing plus PET/CT scanning versus standard testing alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

2

1248

Odds Ratio (M‐H, Random, 95% CI)

1.22 [0.49, 3.04]

Analysis 2.1

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 1 All‐cause mortality.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 1 All‐cause mortality.

2 Cancer‐related mortality Show forest plot

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.55 [0.20, 1.52]

Analysis 2.2

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 2 Cancer‐related mortality.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 2 Cancer‐related mortality.

3 Venous thromboembolism‐related morbidity Show forest plot

1

854

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.48, 2.17]

Analysis 2.3

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 3 Venous thromboembolism‐related morbidity.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 3 Venous thromboembolism‐related morbidity.

4 Characteristics of diagnosed cancer: type of cancer Show forest plot

2

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 4 Characteristics of diagnosed cancer: type of cancer.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 4 Characteristics of diagnosed cancer: type of cancer.

4.1 Acute leukaemia

2

1248

Odds Ratio (M‐H, Random, 95% CI)

1.62 [0.20, 13.22]

4.2 Gynaecological

2

1248

Odds Ratio (M‐H, Random, 95% CI)

2.39 [0.43, 13.36]

4.3 Skin: melanoma

1

854

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.06, 16.34]

4.4 Colorectal

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.43 [0.08, 2.40]

4.5 Prostate

2

1248

Odds Ratio (M‐H, Random, 95% CI)

2.52 [0.48, 13.12]

4.6 Pancreatic

2

1248

Odds Ratio (M‐H, Random, 95% CI)

4.81 [0.55, 42.48]

4.7 Cholangiocarcinoma

1

854

Odds Ratio (M‐H, Random, 95% CI)

0.51 [0.05, 5.63]

4.8 Lymphoma

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.74 [0.09, 5.83]

4.9 Breast

1

854

Odds Ratio (M‐H, Random, 95% CI)

0.20 [0.01, 4.24]

4.10 Urological

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.62 [0.03, 12.32]

4.11 Liver

1

394

Odds Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 8.19]

4.12 Head and neck

1

394

Odds Ratio (M‐H, Random, 95% CI)

3.02 [0.12, 74.47]

4.13 Lung

1

394

Odds Ratio (M‐H, Random, 95% CI)

3.02 [0.12, 74.47]

4.14 Unknown primary

1

854

Odds Ratio (M‐H, Random, 95% CI)

0.34 [0.01, 8.34]

5 Characteristics of diagnosed cancer: stage of cancer Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 5 Characteristics of diagnosed cancer: stage of cancer.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 5 Characteristics of diagnosed cancer: stage of cancer.

5.1 Early

1

394

Odds Ratio (M‐H, Random, 95% CI)

1.78 [0.51, 6.17]

5.2 Advanced

1

394

Odds Ratio (M‐H, Random, 95% CI)

1.0 [0.14, 7.17]

6 Frequency of an underlying cancer diagnosis Show forest plot

2

1248

Odds Ratio (M‐H, Random, 95% CI)

1.71 [0.91, 3.20]

Analysis 2.6

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 6 Frequency of an underlying cancer diagnosis.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 6 Frequency of an underlying cancer diagnosis.

Study flow diagram.
Figures and Tables -
Figure 1

Study flow diagram.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figures and Tables -
Figure 2

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figures and Tables -
Figure 3

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 1 Cancer‐related mortality.
Figures and Tables -
Analysis 1.1

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 1 Cancer‐related mortality.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 2 Characteristics of diagnosed cancer: type of cancer.
Figures and Tables -
Analysis 1.2

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 2 Characteristics of diagnosed cancer: type of cancer.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 3 Characteristics of diagnosed cancer: stage of cancer.
Figures and Tables -
Analysis 1.3

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 3 Characteristics of diagnosed cancer: stage of cancer.

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 4 Frequency of underlying cancer diagnosis.
Figures and Tables -
Analysis 1.4

Comparison 1 Extensive tests versus tests at the physician's discretion, Outcome 4 Frequency of underlying cancer diagnosis.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 1 All‐cause mortality.
Figures and Tables -
Analysis 2.1

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 1 All‐cause mortality.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 2 Cancer‐related mortality.
Figures and Tables -
Analysis 2.2

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 2 Cancer‐related mortality.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 3 Venous thromboembolism‐related morbidity.
Figures and Tables -
Analysis 2.3

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 3 Venous thromboembolism‐related morbidity.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 4 Characteristics of diagnosed cancer: type of cancer.
Figures and Tables -
Analysis 2.4

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 4 Characteristics of diagnosed cancer: type of cancer.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 5 Characteristics of diagnosed cancer: stage of cancer.
Figures and Tables -
Analysis 2.5

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 5 Characteristics of diagnosed cancer: stage of cancer.

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 6 Frequency of an underlying cancer diagnosis.
Figures and Tables -
Analysis 2.6

Comparison 2 Standard testing plus PET/CT scanning versus standard testing alone, Outcome 6 Frequency of an underlying cancer diagnosis.

Summary of findings for the main comparison. Extensive tests versus tests at the physician's discretion

Extensive tests versus tests at the physician's discretion

Patient or population: people with unprovoked VTE

Setting: hospital

Intervention: extensive tests

Comparison: tests at the physician's discretion

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with tests at physician's discretion

Risk with extensive tests

All‐cause mortality1

See comment

See comment

See comment

See comment

See comment

No study measured this outcome.

Cancer‐related mortality2

Study population

OR 0.49
(0.15 to 1.67)

396
(2 RCTs)

⊕⊕⊝⊝
Low3

40 per 1000

20 per 1000
(6 to 65)

VTE‐related mortality4

See comment

See comment

See comment

See comment

See comment

No study measured this outcome.

VTE‐related morbidity5

See comment

See comment

See comment

See comment

See comment

No study measured this outcome.

Stage of cancer ‐ early6

Study population

OR 5.00
(1.05 to 23.76)

201

(1 RCT)

⊕⊕⊝⊝
Low7

20 per 1000

91 per 1000
(21 to 322)

Stage of cancer ‐ advanced8

Study population

OR 0.25
(0.03 to 2.28)

201
(1 RCT)

⊕⊕⊝⊝
Low7

39 per 1000

10 per 1000
(1 to 85)

Time to cancer diagnosis9

See comments

See comments

See comments

201

(1 RCT)

See comments

Time to cancer diagnosis (measured from time of diagnosis of VTE) measured in 1 study (Piccioli 2004b), and reported as a mean of 1 month with extensive tests compared to 11.6 months with tests at physician's discretion (P < 0.001). Standard deviations for these means not given. Attempts to contact author for these data made but no response received.

Frequency of underlying cancer diagnosis10

60 per 1000

78 per 1000
(36 to 158)

OR 1.32
(0.59 to 2.93)

396
(2 RCTs)

⊕⊕⊝⊝
Low3

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; OR: odds ratio; RCT: randomised controlled trial; VTE: venous thromboembolism.

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

1 Death due to any cause.
2 Defined as death due to malignant disease itself, or death due to complications of treatments or procedures to diagnose or treat cancer.
3 Risk of bias was high in two included studies (Piccioli 2004b; Prandoni 2016). Piccioli 2004b terminated early after inclusion of only 201 participants after 5 years for several reasons. First, only five of more than 40 potential participating centres could contribute participants to study. Second, some medical ethics committees rejected the protocol because of absence of screening for occult cancer in the control group, other centres could not start because the proposed extensive screening was judged unethical. Finally, identification of cancer at an apparent early stage in extensive screening group led to an increasing tendency among physicians in participating hospitals to initiate screening for cancer in control participants. Prandoni 2016 study terminated early due to low recruitment rate and failure to show an appreciable advantage of CT‐based strategy over control strategy for detection of cancer.
4 Fatal pulmonary embolism (PE). PE diagnosed "on the basis of a lung scan indicating a high probability of its presence, as indicated by the presence of new or enlarged areas of segmental perfusion defects with ventilation‐perfusion mismatch; an abnormal perfusion scan with documentation of new or recurrent deep vein thrombosis (DVT); the presence of non‐enhancing filling defects in the central pulmonary vasculature on helical computed tomography; a finding of intraluminal filling defects on pulmonary angiography; or evidence of fresh PE at autopsy" (Lee 2003b). Fatal PE including probable fatal PE and unexplained sudden death used if reported, as defined by individual studies.
5 Frequency of recurrent VTE. Recurrent PE or DVT diagnosed if a previously compressible proximal venous segment or segments could no longer be compressed on ultrasonography or if there were constant intraluminal filling defects in two or more projections on venography. Unequivocal extension of the thrombus required for diagnosis of recurrence if results abnormal on previous testing (Lee 2003b)
6 Early‐stage malignancies, defined as T1 or T2 without locoregional or distant metastases (N0 M0).
7 Quality of evidence downgraded for imprecision due to low number of events. Evidence downgraded further as risk of bias high in Piccioli 2004b. Study terminated early after inclusion of only 201 participants after five years for several reasons. First, only five of more than 40 potential participating centres could contribute participants to study. Second, some medical ethics committees rejected the protocol because of absence of screening for occult cancer in the control group, other centres could not start because the proposed extensive screening was judged unethical. Finally, identification of cancer at an apparent early stage in extensive screening group led to an increasing tendency among physicians in participating hospitals to initiate screening for cancer in control participants.
8 Advanced‐stage malignancies, defined as T3 with locoregional or distant metastases (N1 or M1).
9 Time to cancer diagnosis, as defined in included studies.
10 Frequency of an underlying cancer diagnosis (i.e. number of times cancer diagnosed through screening following an unprovoked VTE as defined in included studies) at time of VTE presentation and overall over follow‐up period.

Figures and Tables -
Summary of findings for the main comparison. Extensive tests versus tests at the physician's discretion
Summary of findings 2. Standard testing plus PET/CT scanning versus standard testing alone

Standard testing plus PET/CT scanning versus standard testing alone

Patient or population: people with unprovoked VTE

Setting: hospital

Intervention: standard testing + PET/CT scanning

Comparison: standard testing alone

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

No of participants
(studies)

Quality of the evidence
(GRADE)

Comments

Risk with standard testing alone

Risk with standard testing + PET/CT scanning

All‐cause mortality1

Study population

OR 1.22
(0.49 to 3.04)

1248
(2 RCTs)

⊕⊕⊕⊝
Moderate2

14 per 1000

17 per 1000
(7 to 42)

Cancer‐related mortality3

Study population

OR 0.55
(0.20 to 1.52)

1248
(2 RCTs)

⊕⊕⊕⊝
Moderate2

18 per 1000

10 per 1000
(4 to 26)

VTE‐related mortality4

See comment

See comment

See comment

See comment

See comment

No study measured this outcome.

VTE‐related morbidity5

Study population

OR 1.02
(0.48 to 2.17)

854
(1 RCT)

⊕⊕⊕⊝
Moderate6

32 per 1000

33 per 1000
(16 to 68)

Stage of cancer ‐ early

Study population

OR 1.78
(0.51 to 6.17)

394
(1 RCT)

⊕⊕⊝⊝
Low2,6

20 per 1000

36 per 1000
(10 to 113)

Stage of cancer ‐ advanced

Study population

OR 1.00
(0.14 to 7.17)

394
(1 RCT)

⊕⊕⊝⊝
Low2,6

10 per 1000

10 per 1000
(1 to 69)

Time to cancer diagnosis7

See comments

See comments

See comments

854

(1 RCT)

See comments

Time to cancer diagnosis measured in Carrier 2015 as 4.2 months in standard testing group and 4.0 months in standard testing + PET/CT group (P = 0.88). However, standard deviations for these means not given. Attempts made to contact author for these data but no response received.

Frequency of an underlying cancer diagnosis8

Study population

OR 1.71
(0.91 to 3.20)

1248
(2 RCTs)

⊕⊕⊕⊝
Moderate2

29 per 1000

48 per 1000
(26 to 86)

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; OR: odds ratio; PET/CT: positron emission tomography/computed tomography; RCT: randomised controlled trial; VTE: venous thromboembolism.

GRADE Working Group grades of evidence
High quality: We are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: We are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect.
Very low quality: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

1 Death due to any cause.
2 Quality of evidence downgraded as risk of detection bias high for one study as outcome assessors not blinded to treatment (Robin 2016).
3 Defined as death due to malignant disease itself, or death due to complications of treatments or procedures to diagnose or treat cancer.
4 Fatal pulmonary embolism (PE). PE diagnosed "on the basis of a lung scan indicating a high probability of its presence, as indicated by the presence of new or enlarged areas of segmental perfusion defects with ventilation‐perfusion mismatch; an abnormal perfusion scan with documentation of new or recurrent deep vein thrombosis (DVT); the presence of non‐enhancing filling defects in the central pulmonary vasculature on helical computed tomography; a finding of intraluminal filling defects on pulmonary angiography; or evidence of fresh PE at autopsy" (Lee 2003b). Fatal PE including probable fatal PE and unexplained sudden death used if reported, as defined by individual studies.
5 Frequency of recurrent VTE. Recurrent PE or DVT diagnosed if a previously compressible proximal venous segment or segments could no longer be compressed on ultrasonography or if there were constant intraluminal filling defects in two or more projections on venography.
6 Quality of evidence downgraded for imprecision due to low number of events.
7 Time to cancer diagnosis, as defined in included studies.
8 Frequency of an underlying cancer diagnosis (i.e. number of times cancer diagnosed through screening following an unprovoked VTE as defined in included studies) at time of VTE presentation and overall over follow‐up period.

Figures and Tables -
Summary of findings 2. Standard testing plus PET/CT scanning versus standard testing alone
Comparison 1. Extensive tests versus tests at the physician's discretion

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Cancer‐related mortality Show forest plot

2

396

Odds Ratio (M‐H, Random, 95% CI)

0.49 [0.15, 1.67]

2 Characteristics of diagnosed cancer: type of cancer Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Lung

1

201

Odds Ratio (M‐H, Random, 95% CI)

2.08 [0.19, 23.34]

2.2 Bladder

1

201

Odds Ratio (M‐H, Random, 95% CI)

2.08 [0.19, 23.34]

2.3 Stomach

1

201

Odds Ratio (M‐H, Random, 95% CI)

1.03 [0.06, 16.71]

2.4 Kidney

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.5 Adrenal gland

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.6 Liver

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.7 Uterus

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.8 Breast

1

201

Odds Ratio (M‐H, Random, 95% CI)

1.03 [0.06, 16.71]

2.9 Ovary

1

201

Odds Ratio (M‐H, Random, 95% CI)

3.12 [0.13, 77.55]

2.10 Colon

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.51 [0.05, 5.72]

2.11 Prostate

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.51 [0.05, 5.72]

2.12 Pancreas

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.20 [0.01, 4.26]

3 Characteristics of diagnosed cancer: stage of cancer Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 T1 or T2 (N0 M0)

1

201

Odds Ratio (M‐H, Random, 95% CI)

5.00 [1.05, 23.76]

3.2 T3

1

201

Odds Ratio (M‐H, Random, 95% CI)

0.25 [0.03, 2.28]

4 Frequency of underlying cancer diagnosis Show forest plot

2

396

Odds Ratio (M‐H, Random, 95% CI)

1.32 [0.59, 2.93]

Figures and Tables -
Comparison 1. Extensive tests versus tests at the physician's discretion
Comparison 2. Standard testing plus PET/CT scanning versus standard testing alone

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 All‐cause mortality Show forest plot

2

1248

Odds Ratio (M‐H, Random, 95% CI)

1.22 [0.49, 3.04]

2 Cancer‐related mortality Show forest plot

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.55 [0.20, 1.52]

3 Venous thromboembolism‐related morbidity Show forest plot

1

854

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.48, 2.17]

4 Characteristics of diagnosed cancer: type of cancer Show forest plot

2

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Acute leukaemia

2

1248

Odds Ratio (M‐H, Random, 95% CI)

1.62 [0.20, 13.22]

4.2 Gynaecological

2

1248

Odds Ratio (M‐H, Random, 95% CI)

2.39 [0.43, 13.36]

4.3 Skin: melanoma

1

854

Odds Ratio (M‐H, Random, 95% CI)

1.02 [0.06, 16.34]

4.4 Colorectal

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.43 [0.08, 2.40]

4.5 Prostate

2

1248

Odds Ratio (M‐H, Random, 95% CI)

2.52 [0.48, 13.12]

4.6 Pancreatic

2

1248

Odds Ratio (M‐H, Random, 95% CI)

4.81 [0.55, 42.48]

4.7 Cholangiocarcinoma

1

854

Odds Ratio (M‐H, Random, 95% CI)

0.51 [0.05, 5.63]

4.8 Lymphoma

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.74 [0.09, 5.83]

4.9 Breast

1

854

Odds Ratio (M‐H, Random, 95% CI)

0.20 [0.01, 4.24]

4.10 Urological

2

1248

Odds Ratio (M‐H, Random, 95% CI)

0.62 [0.03, 12.32]

4.11 Liver

1

394

Odds Ratio (M‐H, Random, 95% CI)

0.33 [0.01, 8.19]

4.12 Head and neck

1

394

Odds Ratio (M‐H, Random, 95% CI)

3.02 [0.12, 74.47]

4.13 Lung

1

394

Odds Ratio (M‐H, Random, 95% CI)

3.02 [0.12, 74.47]

4.14 Unknown primary

1

854

Odds Ratio (M‐H, Random, 95% CI)

0.34 [0.01, 8.34]

5 Characteristics of diagnosed cancer: stage of cancer Show forest plot

1

Odds Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 Early

1

394

Odds Ratio (M‐H, Random, 95% CI)

1.78 [0.51, 6.17]

5.2 Advanced

1

394

Odds Ratio (M‐H, Random, 95% CI)

1.0 [0.14, 7.17]

6 Frequency of an underlying cancer diagnosis Show forest plot

2

1248

Odds Ratio (M‐H, Random, 95% CI)

1.71 [0.91, 3.20]

Figures and Tables -
Comparison 2. Standard testing plus PET/CT scanning versus standard testing alone