Scolaris Content Display Scolaris Content Display

Resultados provisionales de la TEP para el pronóstico en adultos con linfoma de Hodgkin: revisión sistemática y metanálisis de los estudios de factores pronósticos

Contraer todo Desplegar todo

Referencias

Andre 2017 {published data only}

Adams HJ, Kwee TC. Interim fluorodeoxyglucose positron emission tomography-adapted therapy is not an efficient approach to improving outcome in early-stage Hodgkin lymphoma. Journal of Clinical Oncology 2017;35(24):2850-1. CENTRAL
Andre ME, Girinsky T, Federico M, Reman O, Fortpied C, Gotti M, et al. Early positron emission tomography response-adapted treatment in stage I and II Hodgkin lymphoma: final results of the randomized EORTC/LYSA/FIL H10 trial (NCT00433433). Journal of Clinical Oncology 2017;35:1786-94. CENTRAL
Andre ME, Reman O, Federico M, Brice P, Brusamolino E, Girinski T, et al. First report on the H10 EORTC/GELA/IIL randomized intergroup trial on early FDG-PET scan guided treatment adaptation versus standard combined modality treatment in patients with supra-diaphragmatic stage I/II Hodgkin's lymphoma, for the Groupe d'Etude Des Lymphomes De l'Adulte (GELA), European Organisation for the Research and Treatment of Cancer (EORTC) Lymphoma Group and the Intergruppo Italiano Linfomi (IIL). Blood 2009;114(22):97. CENTRAL
Andre ME, Reman O, Federico M, Girinski T, Brice P, Brusamolino E, et al. Interim analysis of the randomized EORTC/LYSA/FIL intergroup H10 trial on early PET-scan driven treatment adaptation in stage I/II Hodgkin lymphoma. Blood 2012;120(21):549. CENTRAL
Cottereau AS, Versari A, Loft A, Casanovas O, Bellei M, Ricci R, et al. Prognostic value of baseline metabolic tumor volume in early-stage Hodgkin lymphoma in the standard arm of the H10 trial. Journal of Nuclear Medicine 2018;131(13):1456-63. CENTRAL
Fornecker LM, Lazarovici J, Aurer I, Casasnovas RO, Gac A C, Bonnet C, et al. PET-based response after 2 cycles of brentuximab vedotin in combination with AVD for first-line treatment of unfavorable early-stage Hodgkin lymphoma: First analysis of the primary endpoint of BREACH, a randomized phase II trial of LYSA-FIL-EORTC intergroup. Blood 2017;130(Suppl 1):736. CENTRAL
Hindie E, Mesguich C, Zanotti-Fregonara P. On the role of interim fluorine-18-labeled fluorodeoxyglucose positron emission tomography in early-stage favorable Hodgkin lymphoma. Journal of Clinical Oncology 2017;35(24):2851-2. CENTRAL
Raemaekers JM, Andre MP, Federico M, Girinsky T, Oumedaly R, Brusamolino E, et al. Omitting radiotherapy in early positron emission tomography-negative stage I/II Hodgkin lymphoma is associated with an increased risk of early relapse: Clinical results of the preplanned interim analysis of the randomized EORTC/LYSA/FIL H10 trial. Journal of Clinical Oncology 2014;32:1188-94. CENTRAL

Annunziata 2016 {published data only}

Annunziata S, Calcagni M, Rufini V, Cuccaro A, Massini G, Bartolomei F, et al. The prognostic role of interim FDG-PET/CT and CD68+ cells count in the treatment of Hodgkin lymphoma. European Journal of Nuclear Medicine and Molecular Imaging 2014;41(Suppl. 2):S182. CENTRAL
Annunziata S, Calcagni ML, Indovina L, Rufini V. Measurement uncertainty and clinical impact of target-to-background ratios derived by interim FDG-PET/CT in Hodgkin Lymphoma: reply to Laffon and Martan. European Journal of Nuclear Medicine and Molecular Imaging 2017;44(12):2140-1. CENTRAL
Annunziata S, Cuccaro A, Calcagni M, Indovina L, Hohaus S, Giordano A, et al. Could the ratio between lesion and liver SUVmax (rPET) be a prognostic factor in patients with Hodgkin lymphoma undergoing interim FDG-PET/CT? A retrospective study. In: European Journal of Nuclear Medicine and Molecular Imaging. Vol. 42. 2015:S686. CENTRAL
Annunziata S, Cuccaro A, Calcagni ML, Hohaus S, Giordano A, Rufini V. Interim FDG-PET/CT in Hodgkin lymphoma: the prognostic role of the ratio between target lesion and liver SUVmax (rPET). Annals of Nuclear Medicine 2016;30:588-92. CENTRAL
Annunziata S, Cuccaro A, Hohaus S, Calcagni M L, Giordano A, Rufini V. Interim FDG-PET/CT in patients with Hodgkin lymphoma: The prognostic role of the ratio between target lesion and liver SUVmax (rPET). Journal of Nuclear Medicine 2016;57(Supp 2):651. CENTRAL
Annunziata S, Cuccaro A, Hohaus S, Giordano A, Rufini V. Semi-quantitative parameters could improve positive predictive value of interim FDG-PET/CT in Hodgkin lymphoma. European Journal of Nuclear Medicine and Molecular Imaging 2016;43(Suppl 1):S312. CENTRAL
Annunziata S, Cuccaro A, Hohaus S, Rufini V. Interim FDG-PET/CT in Hodgkin lymphoma: The prognostic value of ratios between target lesion and background SUV. Clinical and Translational Imaging 2017;5(Suppl 1):S44-5. CENTRAL
Annunziata S, Cuccaro A, Rufini V, Calcagni M, Giachelia M, Martini M, et al. The prognostic role of interim FDG-PET/CT, CD68 1 cells count and levels of plasma thymus and activation-regulated chemokine in the treatment of Hodgkin lymphoma. Clinical and Translational Imaging 2015;3(Suppl 1):S18-19. CENTRAL
Cuccaro A, Annunziata S, Rufini V, Calcagni M L, Giordano A, Hohaus S. Can the ratio between lesion and liver SUVmax improve outcome prediction in Hodgkin lymphoma with respect to the 5-point Deauville score? Haematologica 2015;100(Suppl 3):89. CENTRAL

Barnes 2011 {published data only}

Barnes JA, LaCasce AS, Zukotynski K, Israel D, Feng Y, Neuberg D, et al. End-of-treatment but not interim PET scan predicts outcome in nonbulky limited-stage Hodgkin's lymphoma. Annals of Oncology 2011;22:910-5. CENTRAL
Sher DJ, Mauch PM, Van Den Abbeele A, LaCasce AS, Czerminski J, Ng AK. Prognostic significance of mid- and post-ABVD PET imaging in Hodgkin's lymphoma: the importance of involved-field radiotherapy. Annals of Oncology 2009;20:1848-53. CENTRAL

Casasnovas 2019 {published data only}

Casasnovas O, Bouabdallah R, Brice P, Lazarovici J, Ghesquieres H, Stamatoullas A, et al. PET-adapted treatment for newly diagnosed advanced Hodgkin lymphoma (AHL2011): a randomised, multicentre, non-inferiority, phase 3 study. Lancet Oncology 2019;20(2):202-15. CENTRAL
Casasnovas O, Brice P, Bouabdallah R, Salles G, Stamatoulas A, Dupuis J, et al. Final analysis of the AHL2011 randomized phase III LYSA study comparing an early pet driven treatment de-escalation to a not pet-monitored strategy in patients with advanced stages Hodgkin lymphoma. HemaSphere 2018;2(Suppl. 2):7. CENTRAL
Casasnovas O, Brice P, Bouabdallah R, Salles GA, Stamatoulas A, Dupuis J, et al. Randomized phase III study comparing an early PET driven treatment de-escalation to a not PET-monitored strategy in patients with advanced stages Hodgkin lymphoma: final analysis of the AHL2011 LYSA study. Journal of Clinical Oncology 2018;36(15):7503. CENTRAL
Casasnovas O, Brice P, Bouabdallah R, Salles GA, Stamatoullas A, Dupuis J, et al. Randomized phase III study comparing an early pet driven treatment de-escalation to a not pet-monitored strategy in patients with advanced stages hodgkin lymphoma: interim analysis of the AHL2011 LYSA study. Blood 2015;126(23):577. CENTRAL
Casasnovas O, Kanoun S, Tal I, Cottereau AS, Edeline V, Brice P, et al. Baseline total metabolic volume (TMTV) to predict the outcome of patients with advanced Hodgkin lymphoma (HL) enrolled in the AHL2011 LYSA trial. Journal of Clinical Oncology 2016;34(15):7509. CENTRAL
Casasnovas O, Meignan M, Reman O, Gaillard I, Stamatoullas A, Brice P, et al. AHL 2011: A LYSA randomized phase III study of a treatment driven by early PET response compared to a standard treatment in patients with Ann Arbor stage III-IV or high-risk IIB Hodgkin lymphoma. Journal of Clinical Oncology 2013;31(Suppl. 15):8615. CENTRAL

Cerci 2010 {published data only}

Cerci JJ, Pracchia LF, Linardi CC, Pitella FA, Delbeke D, Izaki M, et al. 18F-FDG PET after 2 cycles of ABVD predicts event-free survival in early and advanced Hodgkin lymphoma. Journal of Nuclear Medicine 2010;51:1337-43. CENTRAL

Gallamini 2014 {published data only}

Adams HJ, Kwee TC. Prevention of large-scale implementation of unnecessary and expensive predictive tests in Hodgkin's lymphoma. Lancet Haematology 2017;4(2):e63-4. CENTRAL
Agostinelli C, Gallamini A, Stracqualursi L, Agati P, Tripodo C, Fuligni F, et al. The combined role of biomarkers and interim PET scan in prediction of treatment outcome in classical Hodgkin's lymphoma: a retrospective, European, multicentre cohort study. Lancet Haematology 2016;3(10):e467-79. CENTRAL
Biggi A, Bergesio F, Bianchi A, Menga M, Chauvie S, Fallanca F, et al. Semi-quantitative scan assessment improves the accuracy of the deauville 5-point scale? Clinical and Translational Imaging 2017;5(Suppl. 1):S43-4. CENTRAL
Biggi A, Gallamini A, Chauvie S, Hutchings M, Kostakoglu L, Gregianin M, et al. International validation study for interim PET in ABVD-treated, advanced-stage Hodgkin lymphoma: interpretation criteria and concordance rate among reviewers. Journal of Nuclear Medicine 2013;54:683-90. CENTRAL
Biggi A, Kostakoglu L, Barrington S, Chauvie S, Gregianin M, Meignan M, et al. How the threshold of positive results influence progression free survival (PFS) in advanced Hodgkin's lymphoma treated with ABVD: Experience from the International Validation Study. European Journal of Nuclear Medicine and Molecular Imaging 2012;39(Suppl. 2):S222. CENTRAL
Gallamini A, Barrington SF, Biggi A, Chauvie S, Kostakoglu L, Gregianin M, et al. The predictive role of interim positron emission tomography for Hodgkin lymphoma treatment outcome is confirmed using the interpretation criteria of the Deauville five-point scale. Haematologica 2014;99:1107-13. CENTRAL
Gallamini A, Hutchings M, Rigacci L, Specht L, Merli F, Hansen M, et al. Early interim 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography is prognostically superior to international prognostic score in advanced-stage Hodgkin's lymphoma: a report from a joint Italian-Danish study. Journal of Clinical Oncology 2007;25:3746-52. CENTRAL
Gallamini A, Rigacci L, Merli F, Nassi L, Bosi A, Capodanno I, et al. The predictive value of positron emission tomography scanning performed after two courses of standard therapy on treatment outcome in advanced stage Hodgkin's disease. Haematologica 2006;91:475-81. CENTRAL
Meignan M. High-risk interim PET negative patients in Hodgkin's lymphoma. Lancet Haematology 2016;3(10):e449-50. CENTRAL
Rigacci L, Puccini B, Zinzani PL, Biggi A, Castagnoli A, Merli F, et al. The prognostic value of positron emission tomography performed after two courses (INTERIM-PET) of standard therapy on treatment outcome in early stage Hodgkin lymphoma: A multicentric study by the fondazione italiana linfomi (FIL). American Journal of Hematology 2015;90:499-503. CENTRAL

Gandikota 2015 {published data only}

Gandikota N, Hartridge-Lambert S, Migliacci JC, Yahalom J, Portlock CS, Schoder H. Very low utility of surveillance imaging in early-stage classic Hodgkin lymphoma treated with a combination of doxorubicin, bleomycin, vinblastine, and dacarbazine and radiation therapy. Cancer 2015;121:1985-92. CENTRAL

Hutchings 2005 {published data only}

Hutchings M, Mikhaeel NG, Fields PA, Nunan T, Timothy AR. Prognostic value of interim FDG-PET after two or three cycles of chemotherapy in Hodgkin lymphoma. Annals of Oncology 2005;16:1160-8. CENTRAL

Hutchings 2006 {published data only}

Hutchings M, Loft A, Hansen M, Pedersen LM, Buhl T, Jurlander J, et al. FDG-PET after two cycles of chemotherapy predicts treatment failure and progression-free survival in Hodgkin lymphoma. Blood 2006;107:52-9. CENTRAL

Hutchings 2014 {published data only}

Hutchings M, Kostakoglu L, Zaucha J M, Malkowski B, Biggi A, Danielewicz I, et al. Early determination of treatment sensitivity in Hodgkin lymphoma: FDG-PET/CT after one cycle of therapy has a higher negative predictive value than after two cycles of chemotherapy. Annals of Oncology 2011;22(Suppl. 4):138-9. CENTRAL
Hutchings M, Kostakoglu L, Zaucha JM, Malkowski B, Biggi A, Danielewicz I, et al. In vivo treatment sensitivity testing with positron emission tomography/computed tomography after one cycle of chemotherapy for Hodgkin lymphoma. Journal of Clinical Oncology 2014;32:2705-11. CENTRAL

Kobe 2018 {published data only}

Adams HJ, Kwee TC. Interim FDG-PET does not predict outcome in advanced-stage Hodgkin lymphoma patients treated with BEACOPP. British Journal of Haematology 2018;185(4):758-60. CENTRAL
Adams HJ, Kwee TC. Interim FDG-PET/CT in Hodgkin lymphoma: what are we actually looking at? Acta Oncologica 2018;57(8):1128-30. CENTRAL
Borchmann P, Eichenauer DA, Pluetschow A, Haverkamp H, Kreissl S, Fuchs M, et al. Targeted BEACOPP variants in patients with newly diagnosed advanced stage classical Hodgkin lymphoma: Final analysis of a randomized phase II study. Blood 2015;126(23):580. CENTRAL
Borchmann P, Engert A. Reply to H.J.A. Adams et al, E.A. Hawkes et al, and C.F. Hess et al. Journal of Clinical Oncology 2017;35(3):375-6. CENTRAL
Borchmann P, Goergen H, Kobe C, Eichenauer D, Greil R, Lohri A, et al. EBEACOPP with or without rituximab in interim-PET-positive advanced-stage Hodgkin lymphoma: updated results of the international, randomized phase 3 GHSG HD18 trial. Hematological Oncology 2017;35(Suppl. 2):65. CENTRAL
Borchmann P, Goergen H, Kobe C, Fuchs M, Greil R, Zijlstra JM, et al. Treatment reduction in patients with advanced-stage Hodgkin lymphoma and negative interim pet: final results of the international, randomized phase 3 trial HD18 by the German Hodgkin study group. Haematologica 2017;102(S150):24-5. CENTRAL
Borchmann P, Goergen H, Kobe C, Lohri A, Greil R, Eichenauer DA, et al. Early interim PET in patients with advanced-stage Hodgkin's lymphoma treated within the phase 3 GHSG HD18 study. Blood 2017;130(Suppl. 1):653. CENTRAL
Borchmann P, Goergen H, Kobe C, Lohri A, Greil R, Eichenauer DA, et al. PET-guided treatment in patients with advanced-stage Hodgkin's lymphoma (HD18): final results of an open-label, international, randomised phase 3 trial by the German Hodgkin Study Group. Lancet 2018;390(10114):2790‐802. CENTRAL
Borchmann P, Haverkamp H, Lohri A, Kreissl S, Greil R, Markova J, et al. Addition of rituximab to BEACOPPescalated to improve the outcome of early interim PET positive advanced stage hodgkin lymphoma patients: Second planned interim analysis of the HD18 study. Blood 2014;124(21):500. CENTRAL
Borchmann P, Haverkamp H, Lohri A, Mey U, Kreissl S, Greil R, et al. Progression-free survival of early interim PET-positive patients with advanced stage Hodgkin's lymphoma treated with BEACOPPescalated alone or in combination with rituximab (HD18): an open-label, international, randomised phase 3 study by the German Hodgkin Study Group. Lancet Oncology 2017;18(4):454-63. CENTRAL
Crump M. Predicting outcomes after a positive interim FDG-PET scan in advanced Hodgkin's lymphoma. Lancet Oncology 2017;18(4):416-8. CENTRAL
Kobe C, Goergen H, Baues C, Kuhnert G, Voltin CA, Zijlstra J, et al. Outcome-based interpretation of early interim PET in advanced-stage Hodgkin lymphoma. Blood 2018;132(21):2273-9. CENTRAL
Kobe C, Goergen H, Fuchs M, Eich HT, Baues C, Diehl V, et al. Treatment reduction in patients with advanced-stage Hodgkin lymphoma and negative interim FDG-PET: final results of the international, randomized, phase 3 HD18 trial by the German Hodgkin Study Group. European Journal of Nuclear Medicine and Molecular Imaging 2017;44(Suppl. 1):S312. CENTRAL
Kreissl S, Goergen H, Kobe C, Fuchs M, Greil R, Huttmann A, et al. Treatment reduction in patients with advanced-stage Hodgkin-lymphoma and negative interim PET: final results of the international randomized phase 3 trial HD18 by the German Hodgkin Study Goup. Oncology Research and Treatment 2017;40(Suppl. 3):201. CENTRAL

Markova 2012 {published data only}

Markova J, Kahraman D, Kobe C, Skopalova M, Mocikova H, Klaskova K, et al. Role of [18F]-fluoro-2-deoxy-D-glucose positron emission tomography in early and late therapy assessment of patients with advanced Hodgkin lymphoma treated with bleomycin, etoposide, adriamycin, cyclophosphamide, vincristine, procarbazine and prednisone. Leukemia & Lymphoma 2012;53:64-70. CENTRAL
Markova J, Kobe C, Skopalova M, Dedeckova K, Mocikova H, Klaskova K, et al. Response assessment after 4 cycles of BEACOPP using FDG-PET in patients with advanced-stage Hodgkin lymphoma. Annals of Oncology 2011;22(Suppl. 4):160. CENTRAL
Markova J, Kobe C, Skopalova M, Klaskova K, Dedeckova K, Plutschow A, et al. FDG-PET for assessment of early treatment response after four cycles of chemotherapy in patients with advanced-stage Hodgkin's lymphoma has a high negative predictive value. Annals of Oncology 2009;20:1270-4. CENTRAL
Markova J, Kobe C, Skopalova M, Zikavska L, Vernerova Z, Klaskova K, et al. Early and late response assessment with FDG-PET after BEACOPP-based chemotherapy in advanced-stage Hodgkin lymphoma patients has a high negative predictive value. In: Haematologica. Vol. 94. 2009:33. CENTRAL

Mesguich 2016 {published data only}

Adams HJ, Kwee TC. Hodgkin lymphoma: is there really a need for interim and end-of-treatment FDG-PET evaluations? British Journal of Haematology 2018;181(1):122-3. CENTRAL
Mesguich C, Cazeau AL, Bouabdallah K, Hindie E. Hodgkin lymphoma: is there really a need for interim and end-of-treatment FDG-PET evaluations? - Response to Adams & Kwee. British Journal of Haematology 2018;181(1):124-5. CENTRAL
Mesguich C, Cazeau AL, Bouabdallah K, Soubeyran P, Guyot M, Milpied N, et al. Hodgkin lymphoma: a negative interim-PET cannot circumvent the need for end-of-treatment-PET evaluation. British Journal of Haematology 2016;175:652-60. CENTRAL
Mesguich C, Cazeau AL, Bouabdallah K, Soubeyran P, Guyot M, Milpied N, et al. Hodgkin's lymphoma: Interim FDG-PET result with a score <= 2 on the 5-point scale may obviate the need for end-of-treatment FDG-PET evaluation. Journal of Nuclear Medicine 2015;56(Suppl. 3):595. CENTRAL

Oki 2014 {published data only}

Oki Y, Chuang H, Chasen B, Jessop A, Pan T, Fanale M, et al. The prognostic value of interim positron emission tomography scan in patients with classical Hodgkin lymphoma. British Journal of Haematology 2014;165:112-6. CENTRAL

Okosun 2012 {published data only}

Okosun J, Shaw K, Montoto S, Marcus R, Fields P, Virchis A, et al. Interim FDG-PET scanning in patients with Hodgkin lymphoma and HIV predict response to ABVD chemotherapy. Haematologica 2011;96(Suppl. 2):322-3. CENTRAL
Okosun J, Warbey V, Shaw K, Montoto S, Fields P, Marcus R, et al. Interim fluoro-2-deoxy-D-glucose-PET predicts response and progression-free survival in patients with Hodgkin lymphoma and HIV infection. AIDS 2012;26:861-5. CENTRAL

Orlacchio 2012 {published data only}

Orlacchio A, Schillaci O, Gaspari E, Della Gatta F, Danieli R, Bolacchi F, et al. Role of [18F]-FDG-PET/MDCT in evaluating early response in patients with Hodgkin's lymphoma. La Radiologia Medica 2012;117:1250-63. CENTRAL

Rossi 2014 {published data only}

Rossi C, Kanoun S, Berriolo-Riedinger A, Dygai-Cochet I, Humbert O, Legouge C, et al. Interim 18F-FDG PET SUVmax reduction is superior to visual analysis in predicting outcome early in Hodgkin lymphoma patients. Journal of Nuclear Medicine 2014;55:569-73. CENTRAL

Simon 2016 {published data only}

Barna S, Miltenyi Z, Simon Z, Jona A, Magyari F, Nagy Z, et al. Prognostical value of Interim and restaging PET/CT on Hodgkin lymphoma with CHEAP (Chemotherapy Effectiveness Assessed by PET/CT) long term observation. European Journal of Nuclear Medicine and Molecular Imaging 2014;41(Suppl. 2):S511. CENTRAL
Illes A, Magyari F, Barna S, Simon Z, Payer E, Garai I, et al. Experiences of the first two years with interim PET/CT in Hodgkin lymphoma - the Hungarian CHEAP study. Haematologica 2010;95(Suppl. 4):S45. CENTRAL
Miltenyi Z, Barna S, Garai I, Simon Z, Jona A, Magyari F, et al. Prognostic value of interim and restaging PET/CT in Hodgkin lymphoma. Results of the CHEAP (Chemotherapy Effectiveness Assessment by PET/CT) study - long term observation. Neoplasma 2015;62:627-34. CENTRAL
Miltenyi Z, Simon Z, Jona A, Magyari F, Barna S, Garai I, et al. Interim PET/CT in Hodgkin lymphoma - final results of the Hungarian CHEAP study (2007-2011). Haematologica 2013;98(Suppl. 2):42. CENTRAL
Simon Z, Barna S, Miltenyi Z, Husi K, Magyari F, Jona A, et al. Combined prognostic value of absolute lymphocyte/monocyte ratio in peripheral blood and interim PET/CT results in Hodgkin lymphoma. International Journal of Hematology 2016;103(1):63-9. CENTRAL

Straus 2011 {published data only}

Kostakoglu L, Gandikota N, Hutchings M, Cotter R, Lamonica D, Nanni C, et al. Deauville criteria and post One-Cycle SUVmax decrease seem to predict progression free survival (PFS) better than metabolic tumor measurements in classical Hodgkin lymphoma (cHL). European Journal of Nuclear Medicine and Molecular Imaging 2012;39(Suppl. 2):S222. CENTRAL
Kostakoglu L, Schoder H, Hall N, Straus DJ, Johnson JL, Schwartz L, et al. Interim FDG PET imaging in CALGB 50203 trial of stage I/II non-bulky Hodgkin lymphoma: Would using combined PET and CT criteria better predict response than each test alone? Blood 2011;118(21):3644. CENTRAL
Kostakoglu L, Schoder H, Johnson JL, Hall NC, Schwartz LH, Straus DJ, et al. Interim [(18)F]fluorodeoxyglucose positron emission tomography imaging in stage I-II non-bulky Hodgkin lymphoma: would using combined positron emission tomography and computed tomography criteria better predict response than each test alone? Leukemia & Lymphoma 2012;53:2143-50. CENTRAL
Straus DJ, Johnson JL, LaCasce AS, Bartlett NL, Kostakoglu L, Hsi ED, et al. Doxorubicin, vinblastine, and gemcitabine (CALGB 50203) for stage I/II nonbulky Hodgkin lymphoma: pretreatment prognostic factors and interim PET. Blood 2011;117:5314-20. CENTRAL

Touati 2014 {published data only}

Hutchings M, Kamper P. New clues to the prognostic challenge of Hodgkin Lymphoma. Leukemia & Lymphoma 2015;56(2):277-8. CENTRAL
Touati M, Delage-Corre M, Monteil J, Abraham J, Moreau S, Remenieras L, et al. CD68-positive tumor-associated macrophages predict unfavorable treatment outcomes in classical Hodgkin lymphoma in correlation with interim fluorodeoxyglucose-positron emission tomography assessment. Leukemia and Lymphoma 2015;56:332-41. CENTRAL

Ying 2014 {published data only}

Ying Z, Wang X, Song Y, Zheng W, Wang X, Xie Y, et al. Prognostic value of 18F-FDG PET-CT in Hodgkin lymphoma. Chinese Journal Of Hematology 2014;35:325-7. CENTRAL

Zaucha 2017 {published data only}

Adams HJ, Kwee TC. The predictive value of interim FDG-PET in early-stage Hodgkin lymphoma is not well established. Annals of Oncology 2018;29(2):510-2. CENTRAL
Zaucha JM, Chauvie S, Malkowski B, Warszewska A, Biggi A, Kobylecka M, et al. The prognostic role of interim PET after first chemotherapy cycle in ABVD-treated Hodgkin lymphoma (HL) patients - Polish lymphoma research group (PLRG) observational study. Haematologica 2013;98(Suppl. 2):38. CENTRAL
Zaucha JM, Malkowski B, Chauvie S, Subocz E, Tajer J, Kulikowski W, et al. The predictive role of interim PET after the first chemotherapy cycle and sequential evaluation of response to ABVD in Hodgkin lymphoma patients - the Polish Lymphoma Research Group (PLRG) Observational Study. Annals of Oncology 2017;28(12):3051-7. CENTRAL
Zaucha JM, Malkowski B, Subocz E, Chauvie S, Tajer J, Kulikowski W, et al. The prognostic role of interim PET after first chemotherapy cycle and PET sequential evaluation of response to ABVD in <hodgkin lymphoma patients - The Polish Lymphoma Research Group (PLRG) observational study. Blood 2015;126(23):3943. CENTRAL

Zinzani 2012 {published data only}

Adams HJ, Kwee TC. Does interim 18F-FDG-PET response-adapted therapy really benefit advanced-stage Hodgkin lymphoma patients? Nuclear Medicine Communications 2016;37(12):1333-4. CENTRAL
Puccini B, Rigacci L, Zinzani PL, Broccoli A, Gallamini A, Merli F, et al. Early positive FDG-PET scan do not confirm its prognostic impact in localized bulky disease Hodgkin lymphoma patients. Haematologica 2011;96(Suppl. 3):26-7. CENTRAL
Rigacci L, Puccini B, Gallamini A, Merli F, Stelitano C, Balzarotti M, et al. Early FDG-PET scan confirms its prognostic impact also in localized stage, ABVD treated Hodgkin lymphoma patients. Haematologica 2009;94(Suppl. 2):34. CENTRAL
Rigacci L, Puccini B, Zinzani PL, Kovalchuk S, Broccoli A, Evangelista A, et al. Clinical characteristics of patients with negative interim-PET and positive final PET: Data from the prospective PET-oriented HD0801 study by Fondazione Italiana linfomi (FIL). Hematological Oncology 2017;35(Suppl. 2):38. CENTRAL
Rigacci L, Zinzani PL, Puccini B, Broccoli A, Gallamini A, Merli F, et al. Early FDG-PET scan confirms its prognostic impact also in localized stage, ABVD treated Hodgkin lymphoma patients. Haematologica 2010;95(Suppl. 2):474. CENTRAL
Rigacci L, Zinzani PL, Puccini B, Broccoli A, Gallamini A, Merli F, et al. Early FDG-PET scan confirms its prognostic impact also in localized stage, ABVD treated Hodgkin lymphoma patients. In: Haematologica. Vol. 96. 2010:S13. CENTRAL
Rigacci L, Zinzani PL, Puccini B, Broccoli A, Gallamini A, Merli F, et al. Early positive FDG-PET scan do not confirm its prognostic impact in bulky disease Hodgkin lymphoma patients. Haematologica 2011;96(Suppl. 2):320-1. CENTRAL
Rigacci L, Zinzani PL, Puccini B, Broccoli A, Gallamini A, Merli F, et al. IHP interpretation criteria of interim-PET scan confirms prognostic impact in early stage Hodgkin lymphoma patients without bulky disease. Blood 2010;116(21):3890. CENTRAL
Stefoni V, Broccoli A, Alinari L, Ambrosini V, Derenzini E, Fanti S, et al. Predictive role of early interim FDG-PET in Hodgkin lymphoma. Blood 2009;114(22):1659. CENTRAL
Stefoni V, Pellegrini C, Rigacci L, Broccoli A, Puccini B, Fanti S, et al. Interim PET 2: Is it a real prognostic factor in selecting two different subsets of Hodgkin disease patients? Haematologica 2011;96(Suppl. 3):123. CENTRAL
Zinzani PL, Rigacci L, Stefoni V, Broccoli A, Puccini B, Castagnoli A, et al. Early interim 18F-FDG PET in Hodgkin's lymphoma: evaluation on 304 patients. European Journal of Nuclear Medicine and Molecular Imaging 2012;39:4-12. CENTRAL
Zinzani PL, Tani M, Fanti S, Alinari L, Musuraca G, Marchi E, et al. Early positron emission tomography (PET) restaging: a predictive final response in Hodgkin's disease patients. Annals of Oncology 2006;17:1296-300. CENTRAL

Adams 2016 {published data only}

Adams HJ, Kwee TC. RAPID trial demonstrates low positive predictive value of interim FDG-PET in early-stage Hodgkin lymphoma after three cycles of ABVD. Journal of Pediatric Hematology/Oncology 2016;38(2):165. CENTRAL

Adams 2017 {published data only}

Adams HJ, Kwee TC. Predictive value of interim [18F]fluorodeoxyglucose-positron emission tomography in advanced-stage Hodgkin lymphoma is not well established. Journal of Clinical Oncology2017;35(3):370-1. CENTRAL

Adams 2018 {published data only}

Adams HJ, Kwee TC. Interim FDG-PET has no value in selecting patients who require treatment modification in both early- and advanced-stage Hodgkin lymphoma. British Journal of Haematology2018;183(1):129-31. CENTRAL

Adams 2018a {published data only}

Adams HJ, Kwee TC. No evidence to promote interim FDG-PET adapted therapy in the NCCN guidelines for Hodgkin lymphoma. Journal of the National Comprehensive Cancer Network2018;16(3):226-7. CENTRAL

Adams 2018b {published data only}

Adams HJ, Kwee TC. Strikingly heterogeneous results among studies on interim fluorodeoxyglucose-positron emission tomography-adapted treatment in advanced-stage Hodgkin lymphoma. Journal of Clinical Oncology2018;36(20):2123-4. CENTRAL

Adams 2019 {published data only}

Adams HJ, Kwee TC. Post-ABVD biopsy results, and not post-ABVD FDG-PET results, predict outcome in early-stage Hodgkin lymphoma. British Journal of Haematology2019;184(2):290-2. CENTRAL

Advani 2007 {published data only}

Advani R, Maeda L, Lavori P, Quon A, Hoppe R, Breslin S, et al. Impact of positive positron emission tomography on prediction of freedom from progression after Stanford V chemotherapy in Hodgkin's disease. Journal of Clinical Oncology 2007;25:3902-7. CENTRAL

Afanasyev 2017 {published data only}

Afanasyev BV, Moiseev IS, Alekseev SM, Mikhailova NB, Kondakova EV, Ilyin NV, et al. Multicenter prospective escalation-deescalation PET-guided clinical study in classical type Hodgkin disease in the north-west of Russian federation (RNWOHG-HD1): Rationale and design. Cellular Therapy and Transplantation2017;6(4):76-81. CENTRAL

Albano 2017 {published data only}

Albano D, Patti C, Lagalla R, Midiri M, Galia M. Whole-body MRI, FDG-PET/CT, and bone marrow biopsy, for the assessment of bone marrow involvement in patients with newly diagnosed lymphoma. Journal of Magnetic Resonance Imaging 2017;45(4):1082-9. CENTRAL

Albano 2018 {published data only}

Albano D, Patti C, Matranga D, Lagalla R, Midiri M, Galia M. Whole-body diffusion-weighted MR and FDG-PET/CT in Hodgkin Lymphoma: Predictive role before treatment and early assessment after two courses of ABVD. European Journal of Radiology 2018;103:90-8. CENTRAL

Altamirano 2008 {published data only}

Altamirano J, Esparza JR, de la Garza Salazar J, Calvo PS, Vera SR, Chalapud Revelo JR, et al. Staging, response to therapy, and restaging of lymphomas with18F-FDG PET. Archives of Medical Research 2008;39(1):69-77. CENTRAL

Ansell 2016 {published data only}

Ansell SM. Hodgkin lymphoma: 2016 update on diagnosis, risk-stratification, and management. American Journal of Hematology 2016;91(4):434-42. CENTRAL

Awan 2013 {published data only}

Awan UE, Siddiqui N, SaadUllah M, Bashir H, Farooqui ZS, Muzaffar N, et al. FDG-PET scan in assessing lymphomas and the application of Deauville Criteria. Journal of the Pakistan Medical Association 2013;63(6):725-30. CENTRAL

Barrington 2011a {published data only}

Barrington SF, O'Doherty MJ, Pike L, Franceschetto A, Cucca M, Brun E, et al. Standardised PET-CT reporting for an international multicentre trial in lymphoma (RATHL). European Journal of Nuclear Medicine and Molecular Imaging 2011;38 (SUPPL. 2):S115. CENTRAL

Barrington 2017 {published data only}

Barrington SF, Johnson PW. <18F-FDG PET/CT in lymphoma: Has imaging-directed personalized medicine become a reality? Journal of Nuclear Medicine 2017;58(10):1539-44. CENTRAL

Bar‐Shalom 2003 {published data only}

Bar-Shalom R, Yefremov N, Haim N, Dann EJ, Epelbaum R, Keidar Z, et al. Camera-based FDG pet and 67Ga SPECT in evaluation of lymphoma: Comparative study. Radiology 2003;227(2):353-60. CENTRAL

Basu 2009 {published data only}

Basu S. Early FDG-PET response-adapted risk stratification and further therapeutic decision-making in lymphoma: Will this replace the established prognostic indices and be the standard-of-care in clinical management? European Journal of Nuclear Medicine and Molecular Imaging 2009;36(12):2089-90. CENTRAL

Becherer 2002 {published data only}

Becherer A, Mitterbauer M, Jaeger U, Kalhs P, Greinix HT, Karanikas G, et al. Positron emission tomography with 2-fluoro-D-2-deoxyglucose (FDG-PET) predicts relapse of malignant lymphoma after high-dose therapy with stem cell transplantation. Leukemia 2002;16(2):260-7. CENTRAL

Bednaruk‐Mlynski 2015 {published data only}

Bednaruk-Mlynski E, Pienkowska J, Skorzak A, Malkowski B, Kulikowski W, Subocz E, et al. Comparison of positron emission tomography/computed tomography with classical contrast-enhanced computed tomography in the initial staging of Hodgkin lymphoma. Leukemia and Lymphoma 2015;56(2):377-82. CENTRAL

Biggi 2012 {published data only}

Biggi A, Kostakoglu L, Barrington S, Chauvie S, Gregianin M, Meignan M, et al. How the threshold of positive results influence progression free survival (PFS) in advanced Hodgkin's lymphoma treated with ABVD: Experience from the International Validation Study. European Journal of Nuclear Medicine and Molecular Imaging 2012;39(2):S222. CENTRAL

Biggi 2017 {published data only}

Biggi A, Bergesio F, Menga M, Bianchi A, Fallanca F, Chauvie S, et al. Diagnostic accuracy of FDG PET/CT at the of treatment of Hodgkin lymphoma in the HD0607 trial. Clinical and Translational Imaging 2017;5:S44. CENTRAL

Bishop 2015 {published data only}

Bishop G. PET-directed therapy for Hodgkin's lymphoma. New England Journal of Medicine 2015;373(4):392. CENTRAL

Bjurberg 2006 {published data only}

Bjurberg M, Gustavsson A, Ohlsson T, Brun E. FDG-PET in the detection of residual disease and relapse in patients with Hodgkin's lymphoma. Experience from a Swedish centre. Acta Oncologica 2006;45(6):743-9. CENTRAL

Blum 2002 {published data only}

Blum R, Prince HM, Hicks RJ, Patrikeos A, Seymour J. Discordant response to chemotherapy detected by PET scanning: Unveiling of a second primary cancer. American Journal of Clinical Oncology: Cancer Clinical Trials 2002;25(4):368-70. CENTRAL

Bodet‐Milin 2008 {published data only}

Bodet-Milin C, Kraeber-Bodere F, Moreau P, Campion L, Dupas B, Le Gouill S. Investigation of FDG-PET/CT imaging to guide biopsies in the detection of histological transformation of indolent lymphoma. Haematologica 2008;93(3):471-2. CENTRAL

Bodet‐Milin 2009 {published data only}

Bodet-Milin C, Salaun PY, Crespin C, Vuillez JP, Kraeber-Bodere F. FDG-PET scanning in managing patients with lymphoma. Medecine Nucleaire 2009;33(8):486-90. CENTRAL

Boisson 2007 {published data only}

Boisson N, Cachin F, Kelly A, De Freitas D, Isnardi V, Mestas D, et al. PET-CT in the Hodgkin's disease. Medecine Nucleaire 2007;31(10):562-7. CENTRAL

Borchmann 2016 {published data only}

Borchmann S, von Tresckow B, Engert A. Current developments in the treatment of early-stage classical Hodgkin lymphoma. Current Opinion in Oncology 2016;28(5):377-83. CENTRAL

Bucerius 2006 {published data only}

Bucerius J, Herkel C, Joe AY, Altehoefer C, Finke J, Moser E, et al. <sup>18</sup>F-FDG PET and conventional imaging for assessment of Hodgkin's disease and non Hodgkin's lymphoma: An analysis of 193 patient studies. Nuklearmedizin 2006;45(3):105-10. CENTRAL

Carras 2018 {published data only}

Carras S, Dubois B, Senecal D, Jais JP, Peoc'h M, Quittet P, et al. Interim PET response-adapted strategy in untreated advanced stage Hodgkin lymphoma: Results of GOELAMS LH 2007 phase 2 multicentric trial. Clinical Lymphoma, Myeloma & Leukemia 2018;18(3):191-8. CENTRAL

Ciammella 2016 {published data only}

Ciammella P, Filippi AR, Simontacchi G, Buglione M, Botto B, Mangoni M, et al. Post-ABVD/pre-radiotherapy 18F-FDG-PET provides additional prognostic information for early-stage Hodgkin lymphoma: A retrospective analysis on 165 patients. British Journal of Radiology 2016;89(1061):20150983. CENTRAL

Cremerius 1999 {published data only}

Cremerius U, Fabry U, Kroll U, Zimny M, Neuerburg J, Osieka R, et al. [Clinical value of FDG PET for therapy monitoring of malignant lymphoma--results of a retrospective study in 72 patients]. Clinic for Nuclear Medicine (Stuttgart) 1999;38(1):24-30. CENTRAL

Cremerius 2001 {published data only}

Cremerius U, Fabry U, Neuerburg J, Zimny M, Bares R, Osieka R, et al. Prognostic significance of positron emission tomography using fluorine-18-fluorodeoxyglucose in patients treated for malignant lymphoma. Clinic for Nuclear Medicine (Stuttgart) 2001;40(1):23-30. CENTRAL

Cuccaro 2016 {published data only}

Cuccaro A, Annunziata S, Cupelli E, Martini M, Calcagni ML, Rufini V, et al. CD68+ cell count, early evaluation with PET and plasma TARC levels predict response in Hodgkin lymphoma. Cancer Medicine 2016;5(3):398-406. CENTRAL

D'Urso 2018 {published data only}

D'Urso D, Stefano A, Romano A, Russo G, Cosentino S, Fallanca F, et al. Analysis of metabolic parameters coming from basal and interim PET in Hodgkin lymphoma. Current Medical Imaging Reviews 2018;14(4):533-44. CENTRAL

Damlaj 2017 {published data only}

Damlaj M, Al-Zahrani M, Syed G, Gmati G, Pasha T, Abuelgasim K, et al. Escalation from ABVD following positive interim functional imaging improves progression free survival but not overall survival in advanced classical Hodgkin lymphoma-a real world analysis. Blood 2017;130(Suppl. 1):2799. CENTRAL

Damlaj 2019 {published data only}

Damlaj M, Al-Zahrani M, Syed G, Gmati G, Alahmari B, Pasha T, et al. Interim functional imaging is an independent predictor of progression-free survival in advanced classical Hodgkin lymphoma - A real-world analysis. Clinical Lymphoma, Myeloma and Leukemia 2019;19(1):e71-9. CENTRAL

Danilov 2017 {published data only}

Danilov AV, Li H, Press OW, Shapira I, Swinnen LJ, Noy A, et al. Feasibility of interim positron emission tomography (PET)-adapted therapy in HIV-positive patients with advanced Hodgkin lymphoma (HL): a sub-analysis of SWOG S0816 Phase 2 trial. Leukemia & Lymphoma 2017;58(2):461-5. CENTRAL

Dann 2009 {published data only}

Dann EJ, Bar-Shalom R, Tamir A, Ben-Shachar M, Avivi I, Zuckerman T, et al. For standard and high-risk patients with Hodgkin lymphoma six cycles of tailored BEACOPP, based on interim scintigraphy, are effective and female fertility is preserved. Blood 2009;114(22):1552. CENTRAL

Dann 2010 {published data only}

Dann EJ, Bar-Shalom R, Tamir A, Epelbaum R, Avivi I, Ben-Shachar M, et al. A functional dynamic scoring model to elucidate the significance of post-induction interim fluorine-18-fluorodeoxyglucose positron emission tomography findings in patients with Hodgkin's lymphoma. Haematologica 2010;95(7):1198-206. CENTRAL

Dann 2010a {published data only}

Dann EJ, Bairey O, Bar-Shalom R, Izak M, Korenberg A, Akria L, et al. Tailored therapy in Hodgkin lymphoma, based on predefined risk factors and early interim PET/CT, can lead to modification and safe reduction in therapy: Results of 134 patients on the Israel National Hodgkin Study. Blood 2010;116(21):2809. CENTRAL

Dann 2012 {published data only}

Dann EJ, Bairey O, Bar-Shalom R, Mashiach T, Izak M, Korenberg A, et al. Early Hodgkin lymphoma therapy, based on predefined risk factors and early interim PET/CT. Israeli H2 protocol: Preliminary report. Haematologica 2012;97(Suppl. 1):85. CENTRAL

Dann 2013 {published data only}

Dann EJ, Bairey O, Bar-Shalom R, Izak M, Korenberg A, Akria L, et al. Tailored therapy in Hodgkin lymphoma, based on predefined risk factors and early interim PET/CT, Israeli H2 protocol: Preliminary report on 317 patients. Haematologica 2013;98(Suppl. 2):37. CENTRAL

Dann 2016 {published data only}

Dann EJ, Bairey O, Bar-Shalom R, Mashiach T, Barzilai E, Korenberg A, et al. Adjustment of therapy for Hodgkin lymphoma based on interim PET is beneficial and radiotherapy may be substituted with chemotherapy in patients with negative interim study: Final results of H2 trial. European Haematology Association Open Access Library2016;(S107):4-5. CENTRAL

Dann 2017 {published data only}

Dann EJ, Bairey O, Bar-Shalom R, Mashiach T, Barzilai E, Kornberg A, et al. Modification of initial therapy in early and advanced Hodgkin lymphoma, based on interim PET/CT is beneficial: a prospective multicentre trial of 355 patients. British Journal of Haematology 2017;178:709-718. CENTRAL

Dann 2018 {published data only}

Dann EJ, Paltiel O. Interim FDG-PET has no value in selecting patients who require treatment modification in both early- and advanced-stage Hodgkin lymphoma: Response to Adams and Kwee. British Journal of Haematology 2018;183(1):131-3. CENTRAL

deAndres‐Galiana 2015 {published data only}

deAndres-Galiana EJ, Fernandez-Martinez JL, Luaces O, del Coz JJ, Fernandez R, Solano J, et al. On the prediction of Hodgkin lymphoma treatment response. Clinical and Translational Oncology 2015;17(8):612-9. CENTRAL

Diehl 2007 {published data only}

Diehl V, Kobe C, Haverkamp H, Dietlein M, Engert A. FDG-PET for assessment of residual tissue after completion of chemotherapy in Hodgkin lymphoma report on the 2nd interim analysis of the PET investigation in the trial HD15 of the GHSG. In: Blood. The American Society of Hematology. 2007. CENTRAL

El‐Galaly 2012 {published data only}

El-Galaly TC, Mylam KJ, Brown P, Specht L, Christiansen I, Munksgaard L, et al. Positron emission tomography/computed tomography surveillance in patients with Hodgkin lymphoma in first remission has a low positive predictive value and high costs. Haematologica 2012;97(6):931-6. CENTRAL

Evens 2014 {published data only}

Evens AM, Kostakoglu L. The role of FDG-PET in defining prognosis of Hodgkin lymphoma for early-stage disease. Blood 2014;124(23):3356-64. CENTRAL

Fanti 2008 {published data only}

Fanti S, Castellucci P, Stefoni V, Nanni C, Tani M, Rubello D, et al. Early relapse in a patient with Hodgkin's disease and negative interim FDG-PET. Annals of Nuclear Medicine 2008;22(5):429-32. CENTRAL

Filmont 2003 {published data only}

Filmont J E, Czernin J, Yap C, Silverman DH, Quon A, Phelps ME, et al. Value of F-18 fluorodeoxyglucose positron emission tomography for predicting the clinical outcome of patients with aggressive lymphoma prior to and after autologous stem-cell transplantation. Chest 2003;124(2):608-13. CENTRAL

Fornecker 2017 {published data only}

Fornecker LM, Lazarovici J, Aurer I, Casasnovas RO, Gac AC, Bonnet C, et al. Pet-based response after 2 cycles of brentuximab vedotin in combination with avd for first-line treatment of unfavorable early-stage Hodgkin Lymphoma: first analysis of the primary endpoint of breach, a randomized phase II trial of LYSA-FIL-EORTC intergroup. Blood 2017;130(Suppl. 1):736. CENTRAL

Freudenberg 2004 {published data only}

Freudenberg LS, Antoch G, Schutt P, Beyer T, Jentzen W, Muller SP, et al. FDG-PET/CT in re-staging of patients with lymphoma. European Journal of Nuclear Medicine and Molecular Imaging 2004;31(3):325-9. CENTRAL

Friedberg 2002 {published data only}

Friedberg JW, Fischman A, Neuberg D, Kim H, Takvorian T, Mauch PM, et al. FDG-PET is superior to gallium scintigraphy in the staging and follow-up of patients with de novo Hodgkin's disease: a prospective, blinded comparison. Leukemia & Lymphoma2004;45(1):85-92. CENTRAL

Friedberg 2004 {published data only}

Friedberg JW, Fischman A, Neuberg D, Kim H, Takvorian T, Ng AK, et al. FDG-PET is superior to gallium scintigraphy in staging and more sensitive in the follow-up of patients with de novo Hodgkin lymphoma: a blinded comparison. Leukemia and Lymphoma 2004;45(1):85-92. CENTRAL

Front 1999 {published data only}

Front D, Bar-Shalom R, Mor M, Haim N, Epelbaum R, Frenkel A, et al. Hodgkin disease: prediction of outcome with 67Ga scintigraphy after one cycle of chemotherapy. Radiology 1999;210(2):487-91. CENTRAL

Fruchart 2006 {published data only}

Fruchart C, Reman O, Le Stang N, Musafiri D, Cheze S, Macro M, et al. Prognostic value of early 18 fluorodeoxyglucose positron emission tomography and gallium-67 scintigraphy in aggressive lymphoma: A prospective comparative study. Leukemia and Lymphoma 2006;47(12):2547-57. CENTRAL

Gallamini 2008 {published data only}

Gallamini A, Hutchings M, Avigdor A, Polliack A. Early interim PET scan in Hodgkin lymphoma: Where do we stand? Leukemia and Lymphoma 2008;49(4):659-62. CENTRAL

Gallamini 2017 {published data only}

Gallamini A, Tarella C, Viviani S, Rossi A, Patti C, Mulé A, et al. Early chemotherapy intensification with escalated BEACOPP in advanced-stage Hodgkin lymphoma with a positive interim PET-CT after 2 ABVD cycles: Long-term results of the GITIL/FIL HD 0607 trial. Journal of Clinical Oncology 2018;36(5):454-462. CENTRAL

Gallamini 2018 {published data only}

Gallamini A, Tarella C, Viviani S, Rossi A, Patti C, Mule A, et al. Early chemotherapy intensification with escalated BEACOPP in patients with advanced-stage Hodgkin lymphoma with a positive interim positron emission tomography/computed tomography scan after two ABVD cycles: Long-term results of the GITIL/FIL HD 0607 trial. Journal of Clinical Oncology 2018;36(5):454-62. CENTRAL

Gallamini 2018a {published data only}

Gallamini A, Viviani S, Pavoni C, Rambaldi A. Reply to H.J.A. Adams et al and C. Mesguich et al. Journal of Clinical Oncology 2018;36(20):2127-8. CENTRAL

Gallowitsch 2008 {published data only}

Gallowitsch HJ, Igerc I, Kohlfurst S, Lind P. The incremental value of F-18 FDG PET and PET/CT in malignant lymphoma. Imaging Decisions MRI 2008;12(4):2-6. CENTRAL

Goldschmidt 2011 {published data only}

Goldschmidt N, Or O, Klein M, Savitsky B, Paltiel O. The role of routine imaging procedures in the detection of relapse of patients with Hodgkin lymphoma and aggressive non-Hodgkin lymphoma. Annals of Hematology 2011;90(2):165-71. CENTRAL

Greil 2018 {published data only}

Greil R. Treatment optimisation trial in the first-line treatment of advanced stage Hodgkin lymphoma; comparison of 4-6 cycles of escalated BEACOPP with 4-6 cycles of BrECADD. Memo - Magazine of European Medical Oncology. 2018;11(1):1-28. CENTRAL

Guidez 2016 {published data only}

Guidez S, Delwail V, Brice P. PET-scan in management of Hodgkin's lymphoma. Hematologie 2016;22(6):389-91. CENTRAL

Hagtvedt 2015 {published data only}

Hagtvedt T, Seierstad T, Lund KV, Løndalen AM, Bogsrud TV, Smith HJ, et al. Diffusion-weighted MRI compared to FDG PET/CT for assessment of early treatment response in lymphoma. Acta Radiologica 2015;56(2):152-8. CENTRAL

Haioun 2005 {published data only}

Haioun C, Itti E, Rahmouni A, Brice P, Rain JD, Belhadj K, et al. [18F]fluoro-2-deoxy-D-glucose positron emission tomography (FDG-PET) in aggressive lymphoma: an early prognostic tool for predicting patient outcome. Blood 2005;106(4):1376-81. CENTRAL

Hartmann 2012 {published data only}

Hartmann S, Agostinelli C, Diener J, Doring C, Fanti S, Zinzani PL, et al. GLUT1 expression patterns in different Hodgkin lymphoma subtypes and progressively transformed germinal centers. BMC Cancer 2012;12:586. CENTRAL

Hartridge‐Lambert 2013 {published data only}

Hartridge-Lambert SK, Schoder H, Lim RC, Maragulia JC, Portlock CS. ABVD alone and a PET scan complete remission negates the need for radiologic surveillance in early-stage, nonbulky Hodgkin lymphoma. Cancer 2013;119(6):1203-9. CENTRAL

Honda 2014 {published data only}

Honda A, Nakamura F, Nannya Y, Shintani Y, Fukayama M, Ichikawa M, et al. Pulmonary lymphocyte-rich classical Hodgkin lymphoma with early response to ABVD therapy. Annals of Hematology 2014;93(6):1073-4. CENTRAL

Hueltenschmidt 2001 {published data only}

Hueltenschmidt B, Sautter-Bihl ML, Lang O, Maul FD, Fischer J, Mergenthaler HG, et al. Whole body positron emission tomography in the treatment of Hodgkin disease. Cancer 2001;91(2):302-10. CENTRAL

Huic 2006 {published data only}

Huic D, Mutvar A, Radman I, Grosev D, Labar B, Zuvic M, et al. The value of F-18 FDG triple-head coincidence PET in the posttreatment evaluation of patients with lymphoma. Clinical Nuclear Medicine 2006;31(5):275-8. CENTRAL

Hutchings 2007 {published data only}

Hutchings M, Loft A, Hansen M, Berthelsen AK, Specht L. Clinical impact of FDG-PET/CT in the planning of radiotherapy for early-stage Hodgkin lymphoma. European Journal of Haematology 2007;78(3):206-12. CENTRAL

Iagaru 2008 {published data only}

Iagaru A, Wang Y, Mari C, Quon A, Goris ML, Horning S, et al. (18)F-FDG-PET/CT evaluation of response to treatment in lymphoma: when is the optimal time for the first re-evaluation scan? Hellenic Society of Nuclear Medicine 2008;11(3):153-6. CENTRAL

Illidge 2015 {published data only}

Illidge T. Personalised approach to treating early Hodgkin's lymphoma. BMJ 2015;350:h2927. CENTRAL

Jerusalem 2003 {published data only}

Jerusalem G, Beguin Y, Fassotte MF, Belhocine T, Hustinx R, Rigo P, et al. Early detection of relapse by whole-body positron emission tomography in the follow-up of patients with Hodgkin's disease. Annals of Oncology 2003;14(1):123-30. CENTRAL

Johnson 2015 {published data only}

Johnson PW, Federico M, Fossa A, O'Doherty M, Roberts T, Stevens L, et al. Response-adapted therapy based on interim FDG-PET scans in advanced Hodgkin lymphoma: first analysis of the safety of deescalation and efficacy of escalation in the international RATHL study (CRUK/07/033). Clinical Advances in Hematology and Oncology 2015;13(8 Suppl. 9):6‐7. CENTRAL

Johnson 2016 {published data only}

Johnson P, Federico M, Kirkwood A, Fossa A, Berkahn L, Carella A, et al. Adapted treatment guided by interim PET-CT scan in advanced Hodgkin's lymphoma. New England Journal of Medicine 2016;374(25):2419-29. CENTRAL

Kamran 2016 {published data only}

Kamran SC, Jacene HA, Chen YH, Mauch PM, Ng AK. Clinical outcome of patients with early stage favorable Hodgkin lymphoma treated with ABVDX2 cycles followed by PET/CT restaging and 20 Gy of involved-site radiotherapy. Haematologica 2016;101(Suppl. 5):14. CENTRAL

Kamran 2018 {published data only}

Kamran SC, Jacene HA, Chen YH, Mauch PM, Ng AK. Clinical outcome of patients with early stage favorable Hodgkin lymphoma treated with ABVD x two cycles followed by FDG-PET/CT restaging and 20 Gy of involved-site radiotherapy. Leukemia and Lymphoma 2018;59(6):1384-90. CENTRAL

Kobe 2008 {published data only}

Kobe C, Dietlein M, Franklin J, Markova J, Lohri A, Amthauer H, et al. Positron emission tomography has a high negative predictive value for progression or early relapse for patients with residual disease after first-line chemotherapy in advanced-stage Hodgkin lymphoma. Blood 2008;112(10):3989-94. CENTRAL

Kobe 2014 {published data only}

Kobe C, Kuhnert G, Kahraman D, Haverkamp H, Eich HT, Franke M, et al. Assessment of tumor size reduction improves outcome prediction of positron emission tomography/computed tomography after chemotherapy in advanced-stage Hodgkin lymphoma. Journal of Clinical Oncology 2014;32(17):1776-81. CENTRAL

Kostakoglu 2006 {published data only}

Kostakoglu L, Goldsmith SJ, Leonard JP, Christos P, Furman RR, Atasever T, et al. FDG-PET after 1 cycle of therapy predicts outcome in diffuse large cell lymphoma and classic Hodgkin disease. Cancer 2006;107:2678-87. CENTRAL

Li 2013 {published data only}

Li YJ, Li ZM, Xia XY, Huang HQ, Xia ZJ, Lin TY, et al. Prognostic value of interim and posttherapy 18F-FDG PET/CT in patients with mature T-cell and natural killer cell lymphomas. Journal of Nuclear Medicine 2013;54(4):507-15. CENTRAL

Lowe 2002 {published data only}

Lowe VJ, Wiseman GA. Assessment of Lymphoma Therapy Using (18)F-FDG PET. Journal of Nuclear Medicine 2002;43(8):1028-30. CENTRAL

Milgrom 2017 {published data only}

Milgrom SA, Pinnix CC, Chuang H, Oki Y, Akhtari M, Mawlawi O, et al. Early-stage Hodgkin lymphoma outcomes after combined modality therapy according to the post-chemotherapy 5-point score: can residual pet-positive disease be cured with radiotherapy alone? British Journal of Haematology 2017;179(3):488-96. CENTRAL

Mocikova 2010 {published data only}

Mocikova H, Obrtlikova P, Vackova B, Trneny M. Positron emission tomography at the end of first-line therapy and during follow-up in patients with Hodgkin lymphoma: a retrospective study. Annals of Oncology 2010;21(6):1222-7. CENTRAL

Mocikova 2011 {published data only}

Mocikova H, Pytlik R, Markova J, Steinerova K, Kral Z, Belada D, et al. Pre-transplant positron emission tomography in patients with relapsed Hodgkin lymphoma. Leukemia & Lymphoma 2011;52(9):1668-74. CENTRAL

Molnar 2010 {published data only}

Molnar Z, Simon Z, Borbenyi Z, Deak B, Galuska L, Keresztes K, et al. Prognostic value of FDG-PET in Hodgkin lymphoma for posttreatment evaluation. Long term follow-up results. Neoplasma 2010;57(4):349-54. CENTRAL

Moskowitz 2015 {published data only}

Moskowitz CH. Early FDG-PET adapted treatment improves the outcome of early FDG-PET-positive patients with stages I/II hodgkin lymphoma (HL): Final results of the randomized intergroup EORTC/LYSA/FIL H10 trial. Clinical advances in Hematology & Oncology: H&O 2015;13(8 Supplement 9):16-7. CENTRAL

Naumann 2001 {published data only}

Naumann R, Vaic A, Beuthien-Baumann B, Bredow J, Kropp J, Kittner T, et al. Prognostic value of positron emission tomography in the evaluation of post-treatment residual mass in patients with Hodgkin's disease and non-Hodgkin's lymphoma. British Journal of Haematology 2001;115(4):793-800. CENTRAL

NCT00784537 {published data only}

NCT00784537. High-dose chemotherapy and stem cell transplantation, in patients PET-2 positive, after 2 courses of ABVD and comparison of RT versus no RT in PET-2 negative patients. clinicaltrials.gov/show/nct00784537 (first received 4 November 2008). CENTRAL

NCT00795613 {published data only}

NCT00795613. Positron Emission Tomography (PET)-adapted chemotherapy In advanced Hodgkin Lymphoma (HL). clinicaltrials.gov/show/nct00795613 (first received 21 November 2008). CENTRAL

NCT01358747 {published data only}

NCT01358747. Study of a treatment driven by early PET response to a treatment not monitored by early PET in patients with AA stage 3-4 or 2B HL. clinicaltrials.gov/show/nct01358747 (first received 24 May 2011). CENTRAL

NCT01652261 {published data only}

NCT01652261. Very early FDG-PET/CT-response adapted therapy for advanced Hodgkin Lymphoma (H11). clinicaltrials.gov/show/nct01652261 (first received 30 July 2012). CENTRAL

NCT02292979 {published data only}

NCT02292979. Brentuximab vedotin associated with chemotherapy in untreated patients with Hodgkin Lymphoma. clinicaltrials.gov/show/nct02292979 (first received 18 November 2014). CENTRAL

Nguyen 2017 {published data only}

Nguyen VT, Pophali PA, Tsai J P, Jagadeesh D, Dean RM, Pohlman B, et al. Early stage, bulky Hodgkin lymphoma patients have a favorable outcome when treated with or without consolidative radiotherapy: potential role of PET scan in treatment planning. British Journal of Haematology 2017;179(4):674-6. CENTRAL

Panizo 2004 {published data only}

Panizo C, Perez-Salazar M, Bendandi M, Rodriguez-Calvillo M, Boan JF, Garcia-Velloso MJ, et al. Positron emission tomography using 18F-fluorodeoxyglucose for the evaluation of residual Hodgkin's disease mediastinal masses. Leukemia and Lymphoma 2004;45(9):1829-33. CENTRAL

Paolini 2007 {published data only}

Paolini R, Rampin L, Rodella E, Ramazzina E, Banti E, Al-Nahhas A, et al. The prognostic value of 18F-FDG PET-CT in the management of Hodgkin's lymphoma: Preliminary results of a prospective study. Nuclear Medicine Review 2007;10(2):87-90. CENTRAL

Pavlovsky 2019 {published data only}

Pavlovsky A, Fernandez I, Kurgansky N, Prates V, Zoppegno L, Negri P, et al. PET-adapted therapy after three cycles of ABVD for all stages of Hodgkin lymphoma: results of the GATLA LH-05 trial. British Journal of Haematology 2019;185(5):865-73. CENTRAL

Pichler 2000 {published data only}

Pichler R, Maschek W, Hatzl-Griesenhofer M, Huber H, Wimmer G, Wahl G, et al. Clinical value of FDG hybrid-PET in staging and restaging of malignant lymphoma - compared with conventional diagnostic methods. NuklearMedizin 2000;39(6):166-73. CENTRAL

Reinhardt 2005 {published data only}

Reinhardt MJ, Herkel C, Altehoefer C, Finke J, Moser E. Computed tomography and 18F-FDG positron emission tomography for therapy control of Hodgkin's and non-Hodgkin's lymphoma patients: when do we really need FDG-PET? Annals of Oncology 2005;16(9):1524-9. CENTRAL

Rigacci 2002 {published data only}

Rigacci L, Castagnoli A, Carpaneto A, Carrai V, Vaggelli L, Matteini M. Can (18)F-FDG PET after first cycle chemotherapy predict the efficacy of therapy in Hodgkin's disease? Haematologica 2002;87(5):ELT24. CENTRAL

Rigacci 2017 {published data only}

Rigacci L, Puccini B, Zinzani P, Kovalchuk S, Broccoli A, Evangelista A, et al. Clinical characteristics of patients with negative interim-pet and positive final PET: data from the prospective PET-oriented HD0801 study by Fondazione Italiana linfomi (FIL). Hematological Oncology 2017;35(Suppl. 2):38. CENTRAL

Rubello 2015 {published data only}

Rubello D, Gordien P, Morliere C, Guyot M, Bordenave L, Colletti PM, et al. Variability of hepatic 18F-FDG uptake at interim PET in patients with Hodgkin lymphoma. Clinical Nuclear Medicine 2015;40:e405-10. CENTRAL

Sakr 2017 {published data only}

Sakr R, Massoud M, Kerbage F, Rached L, Zeghondy J, Akoury E, et al. Real-life Experience for Integration of PET-CT in the Treatment of Hodgkin Lymphoma in Lebanon. Clinical Lymphoma, Myeloma & Leukemia 2017;17S:S92-5. CENTRAL

Schot 2007 {published data only}

Schot BW, Zijlstra JM, Sluiter WJ, van Imhoff GW, Pruim J, Vaalburg W, et al. Early FDG-PET assessment in combination with clinical risk scores determines prognosis in recurring lymphoma. Blood 2007;109(2):486-91. CENTRAL

Simontacchi 2015 {published data only}

Simontacchi G, Filippi AR, Ciammella P, Buglione M, Saieva C, Magrini SM, et al. Interim PET after two ABVD cycles in early-stage Hodgkin lymphoma: Outcomes following the continuation of chemotherapy plus radiotherapy. International Journal of Radiation Oncology Biology Physics 2015;92:1077-83. CENTRAL

Slaby 2002 {published data only}

Slaby J, Belohlavek O, Taborska K, Prochazka M, Trneny M, Klener P. [Predictive features of positron emission tomography after two cycles of induction therapy in malignant lymphoma]. Casopis Lékaru Ceských [Journal of Czech Physicians] 2002;141(10):312-5. CENTRAL

Spaepen 2001 {published data only}

Spaepen K, Stroobants S, Dupont P, Thomas J, Vandenberghe P, Balzarini J, et al. Can positron emission tomography with [(18)F]-fluorodeoxyglucose after first-line treatment distinguish Hodgkin's disease patients who need additional therapy from others in whom additional therapy would mean avoidable toxicity? British Journal of Haematology 2001;115:272-8. CENTRAL

Specht 2007 {published data only}

Specht L. FDG-PET scan and treatment planning for early stage Hodgkin lymphoma. Radiotherapy and Oncology 2007;85(2):176-7. CENTRAL

Spinner 2018 {published data only}

Spinner MA, Advani RH. Risk-adapted therapy for advanced-stage Hodgkin lymphoma. Hematology 2018;1:200-6. CENTRAL

Straus 2018 {published data only}

Straus DJ, Jung SH, Pitcher B, Kostakoglu L, Grecula JC, Hsi ED, et al. CALGB 50604: risk-adapted treatment of nonbulky early-stage Hodgkin lymphoma based on interim PET. Blood 2018;132(10):1013-21. CENTRAL

Strigari 2016 {published data only}

Strigari L, Attili A, Duggento A, Chiaravalloti A, Schillaci O, Guerrisi MG. Quantitative analysis of basal and interim PET/CT images for predicting tumor recurrence in patients with Hodgkin's lymphoma. Nuclear Medicine Communications 2016;37:16-22. CENTRAL

Sucak 2011 {published data only}

Sucak GT, Ozkurt ZN, Suyani E, Yasar DG, Akdemir OU, Aki Z, et al. Early post-transplantation positron emission tomography in patients with Hodgkin lymphoma is an independent prognostic factor with an impact on overall survival. Annals of Hematology 2011;90(11):1329-36. CENTRAL

Tirelli 2015 {published data only}

Tirelli U, Spina M. PET-adapted salvage therapy in Hodgkin's lymphoma. Lancet Oncology 2015;16(3):239-40. CENTRAL

Tomita 2015 {published data only}

Tomita N, Hattori Y, Fujisawa S, Hashimoto C, Taguchi J, Takasaki H, et al. Post-therapy 18F-fluorodeoxyglucose positron emission tomography for predicting outcome in patients with peripheral T cell lymphoma. Annals of Hematology 2015;94(3):431-6. CENTRAL

Torizuka 2004 {published data only}

Torizuka T, Nakamura F, Kanno T, Futatsubashi M, Yoshikawa E, Okada H, et al. Early therapy monitoring with FDG-PET in aggressive non-Hodgkin's lymphoma and Hodgkin's lymphoma. European Journal of Nuclear Medicine and Molecular Imaging 2004;31(1):22-8. CENTRAL

Trotman 2017 {published data only}

Trotman J, Fossa A, Federico M, Stevens L, Kirkwood A, Clifton-Hadley L, et al. Response-adjusted therapy for advanced Hodgkin lymphoma (RATHL) trial: longer follow up confirms efficacy of de-escalation after a negative interim PET scan (CRUK/07/033). Hematological Oncology 2017;35:65‐7. CENTRAL

Tseng 2012 {published data only}

Tseng D, Rachakonda LP, Su Z, Advani R, Horning S, Hoppe RT, et al. Interim-treatment quantitative PET parameters predict progression and death among patients with Hodgkin's disease. Radiation Oncology 2012;7:5. CENTRAL

Villa 2018 {published data only}

Villa D, Sehn LH, Aquino-Parsons C, Tonseth P, Scott D W, Gerrie AS, et al. Interim PET-directed therapy in limited stage Hodgkin lymphoma initially treated with ABVD. Haematologica 2018;12:12. CENTRAL

Weidmann 1999 {published data only}

Weidmann E, Baican B, Hertel A, Baum RP, Chow KU, Knupp B, et al. Positron emission tomography (PET) for staging and evaluation of response to treatment in patients with Hodgkin's disease. Leukemia and Lymphoma 1999;34(5-6):545-51. CENTRAL

Wilson 2018 {published data only}

Wilson D, Benard F, Gascoyne RD, Slack GW, Farinha P, Morris J, et al. Interim PET-directed therapy in limited-stage Hodgkin lymphoma initially treated with ABVD. Haematologica 2018;103(12):e590-3. CENTRAL

Xie 2018 {published data only}

Xie W, Jiang X F, Zhao W L, Wang L. Prognostic evaluation of different PET/CT reading methods in Hodgkin lymphoma and diffused large B-cell lymphoma. Journal of Shanghai Jiaotong University 2018;38(8):954-9. CENTRAL

Yasgur 2015 {published data only}

Yasgur BS. Interim PET results guide ongoing therapy in Hodgkin lymphoma. Oncology Report 2015;11(8):available at: https://www.mdedge.com/hematologynews/nhlhub/article/101136/indolent-lymphoma/interim-pet-results-guide-ongoing-therapy. CENTRAL

Yoshimi 2008 {published data only}

Yoshimi A, Izutsu K, Takahashi M, Kako S, Oshima K, Kanda Y, et al. Conventional allogeneic hematopoietic stem cell transplantation for lymphoma may overcome the poor prognosis associated with a positive FDG-PET scan before transplantation. American Journal of Hematology 2008;83(6):477-81. CENTRAL

Zabrocka 2016 {published data only}

Zabrocka E, Sierko E, Wojtukiewicz MZ. Positron emission tomography scanning in the management of Hodgkin lymphoma patients: a single-institution experience. Advances in Clinical and Experimental Medicine 2016;25(6):1185-92. CENTRAL

Zaucha 2009 {published data only}

Zaucha J, Danielewicz I, Malkowski B, Zaucha R, Lesniewski-Kmak K. The role of PET for interim response assessment in patients with Hodgkin's lymphoma. Wspolczesna Onkologia 2009;13(4):161-6. CENTRAL

Zinzani 1999 {published data only}

Zinzani PL, Magagnoli M, Chierichetti F, Zompatori M, Garraffa G, Bendandi M, et al. The role of positron emission tomography (PET) in the management of lymphoma patients. Annals of Oncology 1999;10(10):1181-4. CENTRAL

Zinzani 2002 {published data only}

Zinzani PL, Chierichetti F, Zompatori M, Tani M, Stefoni V, Garraffa G, et al. Advantages of positron emission tomography (PET) with respect to computed tomography in the follow-up of lymphoma patients with abdominal presentation. Leukemia and Lymphoma 2002;43(6):1239-43. CENTRAL

Zinzani 2016 {published data only}

Zinzani PL, Broccoli A, Gioia DM, Castagnoli A, Ciccone G, Evangelista A, et al. Interim positron emission tomography response-adapted therapy in advanced-stage hodgkin lymphoma: final results of the phase II part of the HD0801 study. Journal of Clinical Oncology2016;34(12):1376-85. CENTRAL

Referencias de los estudios en espera de evaluación

Abramson 2010 {published data only}

Abramson JS, Barnes JA, LaCasce AS, Zukotynski K, Israel D, Feng Y, et al. End of treatment but not interim PET scan predicts outcome in non-bulky limited stage Hodgkin lymphoma. Haematologica 2010;95(Suppl. 4):S16. CENTRAL

Algrin 2010 {published data only}

Algrin C, Berenger N, Chevret S, Vercellino L, De Bazelaire C, Brice P, et al. Interim-positron emission tomography with [18F]fluorodeoxyglucose (interim-PET) evaluation in mediastinal lymphoma including Hodgkin lymphoma (HL) and primary mediastinal large B-cell lymphoma (PMBL). Blood 2010;116(21):2860. CENTRAL

Arce‐Calisaya 2013 {published data only}

Arce-Calisaya P, Scarsbrook A, Thygesen H, Chowdhury F, Patel C. Interim FDG PET/CT in Hodgkin's lymphoma - Does binary response assessment criteria have any prognostic value? European Journal of Nuclear Medicine and Molecular Imaging 2013;40(Suppl. 2):S476. CENTRAL

Baratto 2015 {published data only}

Baratto L, Guerra L, Elisei F, Crivellaro C, De Ponti E, Bolis S, et al. Interim-PET in Hodgkin lymphoma: Deauville criteria and metabolic parameters as prognostic factors. Clinical and Translational Imaging 2015;3(Suppl. 1):S16-7. CENTRAL

Barna 2011 {published data only}

Barna S, Fedinecz N, Magyari F, Varga J, Illes A, Garai I. Prognostic value of interim 18FDG-PET/CT in patients with Hodgkin's lymphoma using different 5-point visual scales for interpretation. European Journal of Nuclear Medicine and Molecular Imaging 2011;38(Suppl. 2):S377. CENTRAL

Barrington 2011 {published data only}

Barrington SF, Kostakoglu L, Hutchings M, Meignan M, Biggi A, Gregianin M, et al. Are the Deauville criteria a reliable tool for assessment of interim PET in Hodgkin lymphoma? In: European Journal of Nuclear Medicine and Molecular Imaging. Vol. 38. 2011:S164. CENTRAL

Bentur 2017 {unpublished data only}

Bentur OS, Eldad DJ, Paran E, Lavie D, Nachmias B, Dally N, et al. The predictive value of interim PET-CT in elderly patients with Hodgkin lymphoma. In: Haematologica. Conference: 22th Congress of the European Hematology Association. Spain. Vol. 102. 2017:460-1. CENTRAL

Berenger 2010 {published data only}

Berenger N, Vercellino L, Algrin C, Groheux D, Hindie E, Lussato D, et al. Prognostic value of interim 18F-FDG PET/CT in mediastinal bulky Hodgkin lymphoma. European Journal of Nuclear Medicine and Molecular Imaging 2010;37(Suppl. 2):S436. CENTRAL

Bhatwadekar 2017 {published data only}

Bhatwadekar S, Deshpande S, Khadse S, Shah B, Desai D, Kachchhi U, et al. Excellent outcome in Hodgkin lymphoma with ABVD and CMT: A single-centre retrospective analysis. Hematological Oncology 2017;35(Suppl. 2):316-7. CENTRAL

Cimino 2014 {published data only}

Cimino G, Zaucha JM, Cirillo S, Saviolo C, Hutchings M, El-Galaly TC, et al. The complementary prognostic role of baseline and interim PET in predicting treatment outcome in advanced-stage Hodgkin lymphoma. Blood 2014;124(21):4405. CENTRAL

Cocorocchio 2009 {published data only}

Cocorocchio E, Vanazzi A, Botteri E, Alietti A, Negri M, Bassi S, et al. Prognostic role of interim 18FDG-PET in Hodgkin lymphoma: A single-center experience. Journal of Clinical Oncology 2009;27(15 suppl. 1):e19520. CENTRAL

Cocorocchio 2011 {published data only}

Cocorocchio E, Botteri E, Gigli F, Bassi S, Bertazzoni P, Sammassimo S, et al. Evaluation of interim 18FDG-PET in advanced Hodgkin lymphoma (HL) patients (PTS) treated with ChlVPP/ABVVP regimen. In: Annals of Oncology. Vol. 22. 2011:216. CENTRAL

Copeland 2010 {published data only}

Copeland A, Fanale M, Chuang H, Macapinlac H, Faria SC, Siegmund B, et al. Single institution experience with interim PET evaluation in newly diagnosed CHL receiving ABVD chemotherapy: Need for standardization. Haematologica 2010;95(Suppl. 4):S50. CENTRAL

Cuzzocrea 2015 {published data only}

Cuzzocrea M, Guerra L, Elisei F, Crivellaro C, De Ponti E, Bolis S, et al. The Deauville criteria and metabolic parameters as prognostic factors in interim PET in Hodgkin lymphoma: A single centre experience. European Journal of Nuclear Medicine and Molecular Imaging 2015;42(1 suppl. 1):S152-3. CENTRAL

De Rueda 2013 {published data only}

De Rueda B, Costilla L, Catalina S, Grasa J, Rubio D, Giraldo P. Prognostic value of 18F-FDG PET/CT in Hodgkin lymphoma. Haematologica 2013;98(Suppl. 1):572-3. CENTRAL

Fabbri 2011 {published data only}

Fabbri A, Rigacci L, Lazzi S, Di Lollo S, Pietrini A, Puccini B, et al. 'Early FDG-PET' predicts clinical course of Hodgkin's lymphoma although does not correlate with macrophages infiltration in diagnostic specimens. Haematologica 2011;96(Suppl. 2):321-2. CENTRAL

Fiore 2010 {published data only}

Fiore F, Viviani S, Luminari S, Levis A, Di Raimondo F, Merli F, et al. Early interim FDG-PET during intensified BEACOPP therapy for advanced-stage Hodgkin disease shows a lower positive predictive value than during ABVD. Haematologica 2010;95(Suppl. 4):S19. CENTRAL

Gallegos 2012 {published data only}

Gallegos C, De Rueda B, Grasa JM, Banso A, Giraldo P. The importance of PET/CT as method of evaluation of early response to treatment in HL. Haematologica 2012;97(Suppl. 1):566. CENTRAL

Hohaus 2015 {published data only}

Hohaus S, Cuccaro A, Annunziata S, Martini M, D'Alo F, Calcagni ML, et al. The risk of progression of Hodgkin lymphoma in patients with negative interim PET: A role for the number of tumor-infiltrating macrophages (CD68+ cell counts) and B symptoms. Haematologica 2015;100(Suppl. 3):7. CENTRAL

Hutchings 2010 {published data only}

Hutchings M, Kostakoglu L, Loft A, Coleman M, Specht L. Correlation of FDG-PET results after one cycle and after two cycles of chemotherapy in Hodgkin lymphoma. Journal of Clinical Oncology 2010;28(15):8061. CENTRAL

Knight‐Greenfield 2013 {published data only}

Knight-Greenfield A, Marshall RA, Hutchings M, Doucette J, Stern J, Coleman M, et al. Interim FDG PET/CT to predict progression-free survival (PFS) better than clinical and baseline metabolic measurements in Hodgkin lymphoma (cHL). Journal of Clinical Oncology 2013;31(15):8555. CENTRAL

Leontjeva 2016 {published data only}

Leontjeva A, Demina E, Ryabukhina J, Tumyan G, Trofimova O, Sotnikov V, et al. Significance of early interim PET results in advanced Hodgkin lymphoma treated intensive program EACOPP-14. British Journal of Haematology 2016;173(Suppl. 1):107. CENTRAL

Luminari 2010 {published data only}

Luminari S, Cesaretti M, Tomasello C, Guida A, Bagni B, Merli F, et al. The use of FDG positron emission tomography (FDG-PET) in patients with Hodgkin lymphoma (HL) in the "real world": A population based study from northern Italy. Haematologica 2010;95(Suppl. 4):S42-3. CENTRAL

Luminari 2011 {published data only}NCT0124800010.3109/10428194.2011.580475

Luminari S, Cesaretti M, Tomasello C, Guida A, Bagni B, Merli F, et al. Use of 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography in patients with Hodgkin lymphoma in daily practice: a population-based study from Northern Italy. Leukemia and Lymphoma 2011;52(9):1689-96. CENTRAL

Medvedovskaya 2016 {published data only}

Medvedovskaya E, Leontjeva A, Demina E, Ryabukhina J, Tumyan G, Kokosadze N, et al. The impact of outcome of interim PET/CT on advanced Hodgkin lymphoma treated with EACOPP-14. Haematologica 2016;101(Suppl. 5):26. CENTRAL

Molnar 2011 {published data only}

Molnar Z, Deak B, Kajary K, Lengyel Z, Molnar P, Rosta A, et al. The value of interim 18F-FDG PET/CT in Hodgkin lymphoma. European Journal of Nuclear Medicine and Molecular Imaging 2011;38(Suppl. 2):S165. CENTRAL

Molnar 2011a {published data only}

Molnar Z, Deak B, Kajary K, Lengyel Z, Molnar P, Rosta A, et al. Interim FDG PET/CT examinations in advanced stage Hodgkin lymphoma. Nuclear Medicine Review 2011;14(Suppl. A):A4. CENTRAL

Moreira 2013 {published data only}

Moreira C, Fidalgo P, Queiroz L, Domingues N, Moreira I, Oliveira I, et al. Prognostic value of interim vs. end-of-treatment PET scan in Hodgkin's lymphoma. Hematological Oncology 2013;31(Suppl. 1):257. CENTRAL

Perrone 2009 {published data only}

Perrone T, Gaudio F, Giordano A, De Risi C, Spina A, Curci P, et al. Role of positron emission tomography (PET) after 2 and 4 courses of chemotherapy in patients with Hodgkin's lymphoma: A single center experience. Haematologica 2009;94(Suppl. 4):216. CENTRAL

Pophali 2014 {published data only}

Pophali PA, Rybicki LA, Fenner KB, Jagadeesh D, Dean RM, Pohlman B, et al. Bulky disease does not adversely affect overall survival in early stage Hodgkin lymphoma: Role of interim PET and possible omission of radiotherapy in select patients. Blood 2014;124:21. CENTRAL

Rusconi 2010 {published data only}

Rusconi C, Ravelli E, Gabutti C, Zilioli V, Meli E, Nichelatti M, et al. Baseline and dynamic prognostic factors in newly diagnosed classical Hodgkin's lymphoma. Haematologica 2010;95(Suppl. 4):S43. CENTRAL

Spallino 2017 {published data only}

Spallino M, Guerra L, Cuzzocrea M, Elisei F, Crivellaro C, De Ponti E, et al. The Deauville criteria and QPET as prognostic factors in interim PET in adult Hodgkin lymphoma: A single centre experience. Clinical and Translational Imaging 2017;5(Suppl. 1):S43. CENTRAL

Yaghmour 2012 {published data only}

Yaghmour G, Farhat M, Valdivieso BS, Janakiraman N. PET-negative at 2, 3 or 4 cycles of ABVD in Hodgkin's lymphoma is still good. Journal of General Internal Medicine 2012;27(Suppl. 2):S258-9. CENTRAL

Zanoni 2011 {published data only}

Zanoni L, Agostinelli C, Gallamini A, Rigacci L, Sista M, Piccaluga P, et al. The predictive value of interim PET and immunohistochemical markers in Hodgkin lymphoma (HL). European Journal of Nuclear Medicine and Molecular Imaging 2011;38(Suppl. 2):S165. CENTRAL

NCT00736320 {unpublished data only}

Kobe C, Dietlein M, Kuhnert G, Holstein A, Kahraman D, Haverkamp H, et al. Recruitment and PET interpretation in the HD16 trial for early stage Hodgkin lymphoma. Treatment stratification by FDG-PET. In: NuklearMedizin. Vol. 50. Jahrestagung der Deutschen Gesellschaft für Nuklearmedizin. Bremen, Germany. 2012:A39-40. CENTRAL
NCT00736320. HD16 for early stages - treatment optimization trial in the first-line treatment of early stage Hodgkin lymphoma; treatment stratification by means of FDG-PET. clinicaltrials.gov/ct2/show/nct00736320 (first received 15 August 2008). CENTRAL

Adams 2015a

Adams HJ, Nievelstein RA, Kwee TC. Prognostic value of interim FDG-PET in Hodgkin lymphoma: systematic review and meta-analysis. British Journal of Haematology 2015;170(3):356-66.

Adams 2016a

Adams HJ, Kwee TC. Controversies on the prognostic value of interim FDG-PET in advanced-stage Hodgkin lymphoma. European Journal of Haematology 2016;97(6):491-8.

Altman 1999

Altman DG. Practical Statistics for Medical Research. 1 edition. London: Chapman & Hall, 1991.

Altman 2012

Altman DG, McShane LM, Sauerbrei W, Taube SE. Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK): explanation and elaboration. PLOS Medicine 2012;9(5):e1001216. [PMID: 22675273]

Amitai 2018

Amitai I, Gurion R, Vidal L, Dann EJ, Raanani P, Gafter-Gvili A. PET-adapted therapy for advanced Hodgkin lymphoma – systematic review. Acta Oncologica 2018;57(6):765-72.

Barrington 2014

Barrington SF, Mikhaeel NG, Kostakoglu L, Meignan M, Hutchings M, Mueller SP, et al. Role of imaging in the staging and response assessment of lymphoma: consensus of the International Conference on Malignant Lymphomas Imaging Working Group. Journal of Clinical Oncology 2014;32(27):3048-58.

Barrington 2017a

Barrington SF, Kluge R. FDG PET for therapy monitoring in Hodgkin and non-Hodgkin lymphomas. European Journal of Nuclear Medicine and Molecular Imaging 2017;44(1):97-110.

Berriolo‐Riedinger 2018

Berriolo-Riedinger A, Becker S, Casasnovas O, Vander Borght T, Edeline V. Role of FDG PET-CT in the treatment management of Hodgkin lymphoma. Cancer Radiotherapy 2018;22(5):393-400.

Blank 2017

Blank O, von Tresckow B, Monsef I, Specht L, Engert A, Skoetz N. Chemotherapy alone versus chemotherapy plus radiotherapy for adults with early stage Hodgkin lymphoma. Cochrane Database of Systematic Reviews 2017, Issue 4. Art. No: CD007110. [DOI: 10.1002/14651858.CD007110.pub3]

Boellaard 2010

Boellaard R, O'Doherty MJ, Weber WA, Mottaghy FM, Lonsdale MN, Stroobants SG, et al. FDG PET and PET/CT: EANM procedure guidelines for tumour PET imaging: version 1.0. European Journal of Nuclear Medicine and Molecular Imaging 2010;37(1):181-200. [PMID: 19915839]

Borchmann 2011

Borchmann P, Haverkamp H, Diehl V, Cerny T, Markova J, Ho AD, et al. Eight cycles of escalated-dose BEACOPP compared with four cycles of escalated-dose BEACOPP followed by four cycles of baseline-dose BEACOPP with or without radiotherapy in patients with advanced-stage Hodgkin's lymphoma: final analysis of the HD12 trial of the German Hodgkin Study Group. Journal of Clinical Oncology 2011;29(32):4234-42. [PMID: 21990399]

Bouwmeester 2012

Bouwmeester W, Zuithoff NP, Mallett S, Geerlings MI, Vergouwe Y, Steyerberg EW, et al. Reporting and methods in clinical prediction research: a systematic review. PLOS Medicine 2012;9(5):1-12.

Bröckelmann 2018

Bröckelmann PJ, Eichenauer DA, Jakob T, Follmann M, Engert A, Skoetz N. Clinical practice guideline: Hodgkin lymphoma in adults—diagnosis, treatment, and follow-up. Deutsches Ärzteblatt International 2018;115:535-40.

Canellos 1992

Canellos GP, Anderson JR, Propert KJ, Nissen N, Cooper MR, Henderson ES, et al. Chemotherapy of advanced Hodgkin's disease with MOPP, ABVD, or MOPP alternating with ABVD. New England Journal of Medicine 1992;327(21):1478-84. [PMID: 1383821]

Cheson 2014

Cheson BD, Fisher RI, Barrington SF, Cavalli F, Schwartz LH, Zucca E, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. Journal of Clinical Oncology 2014;32(27):3059-68. [PMID: 25113753]

Covidence [Computer program]

Veritas Health InnovationCovidence. Version accessed 4 March 2019. Melbourne, Australia.: Veritas Health Innovation. Available at www.covidence.org.

Cuccaro 2014

Cuccaro A, Bartolomei F, Cupelli E, Galli E, Giachelia M, Hohaus S. Prognostic factors in hodgkin lymphoma. Mediterranean Journal of Hematology and Infectious Diseases 2014;6(1):e2014053.

Debray 2017

Debray T, Damen J, Snell K, Ensor J, Hooft L, Reitsma J, et al. A guide to systematic review and meta-analysis of prediction model performance. BMJ 2017;356:i6460.

Debray 2018

Debray T, Moons K, Riley R. Detecting small-study effects and funnel plot asymmetry in meta-analysis of survival data: A comparison of new and existing tests. Research Synthesis Methods 2018;9(1):41-50.

Deeks 2011

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

Engert 2003

Engert A, Schiller P, Josting A, Herrmann R, Koch P, Sieber M, et al. Involved-field radiotherapy is equally effective and less toxic compared with extended-field radiotherapy after four cycles of chemotherapy in patients with early-stage unfavorable Hodgkin's lymphoma: results of the HD8 trial of the German Hodgkin's Lymphoma Study Group. Journal of Clinical Oncology 2003;21(19):3601-8.

Engert 2007

Engert A, Franklin J, Eich HT, Brillant C, Sehlen S, Cartoni C, et al. Two cycles of doxorubicin, bleomycin, vinblastine, and dacarbazine plus extended-field radiotherapy is superior to radiotherapy alone in early favorable Hodgkin's lymphoma: final results of the GHSG HD7 trial. Journal of Clinical Oncology 2007;25(23):3495-502.

Engert 2010

Engert A, Plutschow A, Eich HT, Lohri A, Dorken B, Borchmann P, et al. Reduced treatment intensity in patients with early-stage Hodgkin's lymphoma. New England Journal of Medicine 2010;363(7):640-52. [PMID: 20818855]

Engert 2012

Engert A, Haverkamp H, Kobe C, Markova J, Renner C, Ho A, et al. Reduced-intensity chemotherapy and PET-guided radiotherapy in patients with advanced stage Hodgkin's lymphoma (HD15 trial): a randomised, open-label, phase 3 non-inferiority trial. Lancet 2012;379(9828):1791-9. [PMID: 22480758]

Fitzgerald 2019

Fitzgerald TJ. Optimisation of adaptive therapy for advanced Hodgkin lymphoma. Lancet Oncology 2019;20(2):167-8.

Franklin 2005

Franklin JG, Paus MD, Pluetschow A, Specht L. Chemotherapy, radiotherapy and combined modality for Hodgkin's disease, with emphasis on second cancer risk. Cochrane Database of Systematic Reviews 2005, Issue 4. Art. No: CD003187. [DOI: 10.1002/14651858.CD003187.pub2] [PMID: 16235316]

Gallamini 2007

Gallamini A, Hutchings M, Rigacci L, Specht L, Merli F, Hansen M, et al. Early interim 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography is prognostically superior to international prognostic score in advanced-stage Hodgkin's lymphoma: a report from a joint Italian-Danish study. Journal of Clinical Oncology 2007;25(24):3746-52. [PMID: 17646666]

Geersing 2012

Geersing GJ, Bouwmeester W, Zuithoff P, Spijker R, Leeflang M, Moons KG. Search filters for finding prognostic and diagnostic prediction studies in Medline to enhance systematic reviews. PLOS One 2012;7(7):e32844.

Hayden 2013

Hayden JA, van der Windt DA, Cartwright JL, Cote P, Bombardier C. Assessing bias in studies of prognostic factors. Annals of Internal Medicine 2013;158(4):280-6. [PMID: 23420236]

Higgins 2011

Higgins JP, Deeks JJ (editors). Chapter 7: Selecting studies and collecting data. In: Higgins JP, Green S (editors), Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from www.cochrane-handbook.org.

Higgins 2011b

Higgins JP, Deeks JJ, Altman DG. Chapter 16: Special topics in statistics. In: Higgins JP, Green S, editors(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011.

Howlader 2015

Howlader N, Noone AM, Krapcho M, Garshell J, Neyman N, Altekruse SF, et al. SEER Cancer Statistics Review, 1975-2010. National Cancer Institute. Bethesda, MD. http://seer.cancer.gov/csr/1975_2010/ based on November 2012 SEER data submission, posted to the SEER web site, April 2013 (accessed 23 June 2015).

Iorio 2015

Iorio A, Spencer FA, Falavigna M, Alba C, Lang E, Burnand B, et al. Use of GRADE for assessment of evidence about prognosis: rating confidence in estimates of event rates in broad categories of patients. BMJ 2015;350:1-8.

Josting 2010

Josting A. Prognostic factors in Hodgkin lymphoma. Expert Review of Hematology 2010;3(5):583-92.

Juweid 2007

Juweid ME, Stroobants S, Hoekstra OS, Mottaghy FM, Dietlein M, Guermazi A, et al. Use of positron emission tomography for response assessment of lymphoma: consensus of the Imaging Subcommittee of International Harmonization Project in Lymphoma. Journal of Clinical Oncology 2007;25:571-8.

Klimm 2005

Klimm B, Engert A, Diehl V. First-line treatment of Hodgkin's lymphoma. Current Hematology Reports 2005;4(1):15-22.

Kobe 2008a

Kobe C, Dietlein M, Franklin J, Markova J, Lohri A, Amthauer H, et al. Positron emission tomography has a high negative predictive value for progression or early relapse for patients with residual disease after first-line chemotherapy in advanced-stage Hodgkin lymphoma. Blood 2008;112(10):3989-94. [PMID: 18757777]

Kobe 2010

Kobe C, Dietlein M, Fuchs M. Interpretation and validation of interim positron emission tomography in Hodgkin lymphoma. Leukemia and Lymphoma2010;51(3):552-3. [PMID: 20141443]

Kobe 2010a

Kobe C, Dietlein M, Kriz J, Furth C, Fuchs M, Borchmann P, et al. The role of PET in Hodgkin's lymphoma and its impact on radiation oncology. Expert Review of Anticancer Therapy 2010;10(9):1419-28. [PMID: 20836677]

Kyzas 2005

Kyzas PA, Loizou KT, Ioannidis JP. Selective reporting biases in cancer prognostic factor studies. Journal of the National Cancer Institute 2005;97(14):1043-55. [PMID: 16030302]

Kyzas 2007

Kyzas PA, Denaxa-Kyza D, Ioannidis JP. Quality of reporting of cancer prognostic marker studies: association with reported prognostic effect. Journal of the National Cancer Institute 2007;99(3):236-43. [PMID: 17284718]

Kılıçkap 2013

Kılıçkap S, Barışta I, Ulger S, Celik I, Selek U, Yıldız F, et al. Clinical Features and Prognostic Factors of Hodgkin's Lymphoma: A Single Center Experience. Balkan Medical Journal 2013;30(2):178-85.

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for studies. In: Higgins JP, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from www.cochrane-handbook.org.

Lister 1989

Lister TA, Crowther D, Sutcliffe SB, Glatstein E, Canellos GP, Young RC, et al. Report of a committee convened to discuss the evaluation and staging of patients with Hodgkin's disease: Cotswolds meeting. Journal of Clinical Oncology 1989;7(11):1630-6.

Macaskill 2010

Macaskill P, Gatsonis C, Deeks J, Harbord R, Takwoingi Y. Chapter 10 Analysing and Presenting Results. In: Deeks JJ, Bossuyt PM, Gatsonis C, editors(s). Cochrane Handbook for Systematic Reviews of Diagnostic Test Accuracy. Version 1.0. The Cochrane Collaboration, 2010. Available from: http://srdta.cochrane.org/.

Mallett 2010

Mallett S, Timmer A, Sauerbrei W, Altman DG. Reporting of prognostic studies of tumour markers: a review of published articles in relation to REMARK guidelines. British Journal of Cancer 2010;102(1):173-80. [PMID: 19997101]

Markova 2009

Markova J, Kobe C, Skopalova M, Klaskova K, Dedeckova K, Plutschow A, et al. FDG-PET for assessment of early treatment response after four cycles of chemotherapy in patients with advanced-stage Hodgkin's lymphoma has a high negative predictive value. Annals of Oncology 2009;20(7):1270-4. [PMID: 19228806]

McShane 2005

McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Reporting recommendations for tumour MARKer prognostic studies (REMARK). British Journal of Cancer 2005;93(4):387-91.

Meignan 2009

Meignan M, Gallamini A, Meignan M, Gallamini A, Haioun C. Report on the First International Workshop on Interim-PET-Scan in Lymphoma. Leukemia and Lymphoma 2009;50(8):1257-60.

Meignan 2009a

Meignan M, Itti E, Bardet S, Lumbroso J, Edeline V, Olivier P, et al. Development and application of a real-time on-line blinded independent central review of interim PET scans to determine treatment allocation in lymphoma trials. Journal of Clinical Oncology 2009;27(16):2739-41.

Moher 2009

Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Journal of Clinical Epidemiology 2009;62(10):1006-12.

Moons 2009

Moons KG, Royston P, Vergouwe Y, Grobbee DE, Altman DG. Prognosis and prognostic research: what, why, and how? BMJ 2009;338:b375.

Moons 2014

Moons KG, de Groot JA, Bouwmeester W, Vergouwe Y, Mallett S, Altman DG, et al. Critical appraisal and data extraction for systematic reviews of prediction modelling studies: the CHARMS checklist. PLoS Medicine 2014;11:e1001744.

Moskowitz 2018

Moskowitz CH, Walewski J, Nademanee A, Masszi T, Agura E, Holowiecki J, et al. Five-year PFS from the AETHERA trial of brentuximab vedotin for Hodgkin lymphoma at high risk of progression or relapse. Blood Journal 2018;132(25):2639.

Parmar 1998

Parmar MK, Torri V, Stewart L. Extracting summary statistics to perform meta-analyses of the published literature for survival endpoints. Statistics in Medicine 1998;17(24):2815-34.

Peat 2014

Peat G, Riley RD, Croft P, Morley KI, Kyzas PA, Moons KG, et al. Improving the transparency of prognosis research: the role of reporting, data sharing, registration, and protocols. PLOS Medicine 2014;11(7):e1001671.

Radford 2015

Radford J, Illidge T, Counsell N, Hancock B, Pettengell R, Johnson P, et al. Results of a trial of PET-directed therapy for early-stage Hodgkin's lymphoma. New England Journal of Medicine 2015;372(1):1598-607.

Rancea 2013

Rancea M, Monsef I, von Tresckow B, Engert A, Skoetz N. High-dose chemotherapy followed by autologous stem cell transplantation for patients with relapsed/refractory Hodgkin lymphoma. Cochrane Database of Systematic Reviews 2013, Issue 6. Art. No: CD009411. [DOI: 10.1002/14651858.CD009411.pub2] [PMID: 23784872]

Rancea 2013a

Rancea M, Engert A, von Tresckow B, Halbsguth T, Behringer K, Skoetz N. Hodgkin's lymphoma in adults: diagnosis, treatment and follow-up. Deutsches Arzteblatt International 2013;110(11):177-83, 183e1-3. [PMID: 23555321]

Re 2005

Re D, Thomas RK, Behringer K, Diehl V. From Hodgkin disease to Hodgkin lymphoma: biologic insights and therapeutic potential. Blood 2005;105(12):4553-60. [PMID: 15728122]

Review Manager 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane CollaborationReview Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Riley 2013

Riley RD, Hayden JA, Steyerberg EW, Moons KG, Abrams K, Kyzas PA, et al. Prognosis Research Strategy (PROGRESS) 2: prognostic factor research. PLOS Medicine 2013;10(2):e1001380. [PMID: 23393429]

Riley 2019

Riley RD, Moons KG, Snell KE, Ensor J, Hooft L, Altman DG, et al. A guide to systematic review and meta-analysis of prognostic factor studies. BMJ 2019;364:k4597.

Riley 2019b

Riley RD, van der Windt D, Croft P, Moons KGM. Prognosis Research in Healthcare. Concepts, Methods, and Impact. Oxford University Press, 2019.

Sauerbrei 2005

Sauerbrei W. Prognostic factors. Confusion caused by bad quality design, analysis and reporting of many studies. Advances in Oto-Rhino-Laryngology 2005;62:184-200. [PMID: 15608428]

Sickinger 2015

Sickinger MT, von Tresckow B, Kobe C, Engert A, Borchmann P, Skoetz N. Positron emission tomography-adapted therapy for first-line treatment in individuals with Hodgkin lymphoma. Cochrane Database of Systematic Reviews 2015, Issue 1. Art. No: CD010533. [DOI: 10.1002/14651858.CD010533.pub2]

Skoetz 2013

Skoetz N, Trelle S, Rancea M, Haverkamp H, Diehl V, Engert A, et al. Effect of initial treatment strategy on survival of patients with advanced-stage Hodgkin's lymphoma: a systematic review and network meta-analysis. Lancet. Oncology 2013;14(10):943-52. [PMID: 23948348]

Skoetz 2017a

Skoetz N, Will A, Monsef I, Brillant C, Engert A, von Tresckow B. Comparison of first-line chemotherapy including escalated BEACOPP versus chemotherapy including ABVD for people with early unfavourable or advanced stage Hodgkin lymphoma. Cochrane Database of Systematic Reviews 2017, Issue 5. Art. No: CD007941. [DOI: 10.1002/14651858.CD007941.pub3]

Steyerberg 2013

Steyerberg EW, Moons KG, van der Windt DA, Hayden JA, Perel P, Schroter S, et al. Prognosis Research Strategy (PROGRESS) 3: prognostic model research. PLOS Medicine 2013;10(2):e1001381.

Swerdlow 2008

Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO Classification of Tumours of the Haematopoietic and Lymphoid Tissues. Fourth edition. Vol. 2. Lyon: WHO Press, 1211 Geneva 27, Switzerland, 2008. [ISBN-13: 9789283224310]

Thomas 2002

Thomas RK, Re D, Zander T, Wolf J, Diehl V. Epidemiology and etiology of Hodgkin's lymphoma. Annals of Oncology 2002;13:147-52. [PMID: 12401681]

Tierney 2007

Tierney JF, Stewart LA, Ghersi D, Burdett S, Sydes MR. Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 2007;8:16.

Trivella 2006

Trivella M. Systematic Reviews of Prognostic Factor Studies (Section: Estimating the Hazard Ratio) [DPhil]. University of Oxford (Altman DG, thesis advisor) June 10th 2006:219.

von Tresckow 2012

von Tresckow B, Plutschow A, Fuchs M, Klimm B, Markova J, Lohri A, et al. Dose-intensification in early unfavorable Hodgkin's lymphoma: final analysis of the German Hodgkin study group HD14 trial. Journal of Clinical Oncology 2012;30(9):907-13. [PMID: 22271480]

Weigler‐Sagie 2010

Weiler-Sagie M, Bushelev O, Epelbaum R, Dann EJ, Haim N, Avivi I, et al. (18)F-FDG avidity in lymphoma readdressed: a study of 766 patients. Journal of Nuclear Medicine 2010;51(1):25-30.

Referencias de otras versiones publicadas de esta revisión

Skoetz 2017

Skoetz N, Collins G, Moons K, Estcourt LJ, Engert A, Kobe C, et al. Interim PET for prognosis in adults with Hodgkin lymphoma: a prognostic factor exemplar review. Cochrane Database of Systematic Reviews 2017, Issue 4. Art. No: CD012643. [DOI: 10.1002/14651858.CD012643]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Andre 2017

Study characteristics

Methods

Secondary citation(s)

  • Raemarkers 2014, Cottereau 2018

Language of publication

  • English

Study design

  • Prospective, multi‐centre, phase III randomised trial

Study centre(s)

  • Various

Countries

  • Belgium, Croatia, Denmark, France, Italy, the Netherlands, Slovakia, Switzerland

Median follow‐up time (range)

  • 55 months

Participants

Number of included participants

  • Total: 1925

  • Randomised to standard treatment without change in protocol because of interim PET: 954

Inclusion criteria

  • Previously untreated

  • Classic supradiaphragmic stage I and II HL

  • Age 15 to 70 years

Exclusion criteria

  • Previous laparotomy

  • Concomitant or previous cancer other than basal‐cell carcinoma of the skin or in situ carcinoma of the cervix

  • Concomitant severe illness that would reduce life expectancy

  • Social circumstances not allowing for proper treatment and follow‐up

  • Positivity for the human immunodeficiency virus

(exclusion criteria reported in Fermé 20071)

Consent

  • Yes; written informed consent

Recruitment period

  • November 2006 to June 2011

Age (range, in years)

  • Favourable, standard treatment group median: 31 (15‐49)

  • Unfavourable, standard treatment group median: 32 (15‐70)

Ethnic group(s)

  • Not reported

Stages of disease

  • Early stages (I and II)

Comorbidities

  • Not reported, except for the exclusion criteria

Therapy regimen

  • ABVD and radiotherapy depending on treatment arm, favourable/unfavourable disease, and early PET (ePET) positivity

Prognostic factor(s)

Prognostic factor(s)

  • Early PET (ePET)

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • After 2 ABVD cycles

Method for measurement (use of specific scale and cut‐off)

  • International Harmonization Project criteria. According to these criteria: PET‐negative corresponds to Deauville score 1 (no uptake) and score 2 (uptake ≤ mediastinum)

  • Central review performed online (up to 6 experts, and one local expert)

Was the same definition and method for measurement used in all participants?

  • Central review started later for 2 centres in Italy due to technical difficulties, only 75% of ePET were centrally reviewed

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as time from random assignment to date of progression (as experiencing relapse after previous complete remission, progressive disease, or death from any cause)

Secondary outcome(s) and definition(s)

  • Overall survival (OS), not defined

Timing of outcome measurement

  • At 5 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included inunivariateanalysis for each outcome

  • PFS: all

  • OS: all

Statistical method

  • Kaplan‐Meier method

  • HR (95% CI)

  • Randomised arms were compared using the log‐rank test stratified by Ann Arbor stage and availability of a baseline FDG‐PET scan

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • Length of follow‐up reported. Exclusion of participants due to safety amendment during the study.

Prognostic factor measurement

  • Moderate risk

  • Adequate measurement and description. Central review only for 75% of scans and delayed in the case of 2 centres due to technical difficulties.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • No definition of outcome. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • Casasnovas O: honoraria received from Genentech, Takeda, Gilead Sciences, Sanofi; consulting or advisory role at Genentech, Takeda, Gilead Sciences; research funding received from Genentech; travel, accomodation, expenses received from Genentech, Takeda, Gilead Sciences

  • Brice P: research funding received from Merck Sharp & Dohme Oncology, Takeda; travel, accomodation, expenses received from Takeda

  • Specht L: consulting or advisory role at Takeda; research funding received from Varian Medical Systems; travel, accomodation, expenses received from Takeda

  • Delarue R: honoraria received from Servier, Gilead Sciences, Roche, Celgene, Takeda; consulting or advisory role at Gilead Sciences, Roche; Speakers' Bureau at Karyopharm Therapeutics; travel, accomodation, expenses received from Roche, Takeda, Celgene

  • Hutchings M: consulting or advisory role at Takeda, Genentech, Celgene, Bayer; research funding received from Takeda, Janssen‐Cilag, Genentech, Celgene; travel, accomodation, expenses received from Takeda, Bristol‐Myers, Squibb, Janssen‐Cilag

Funding

  • Supported by European Organisation for Research and Treatment of Cancer (Belgium), LYmphoma Study Association (France), Fondazione Italiana Limfomi (Italy), Fondation Belge Contre le Cancer (Belgium), Dutch Cancer Society (the Netherlands), Institut National du Cancer (France), Assistance Publique des Hopitaux de Paris (France), Societe Française de Medecine Nucleaire et Imagerie Moleculaire (France), Associazone Angela Serra (Italy), van Vlissingen Lymfoom Fonds (the Netherlands), and Chugai Pharmaceutical (Japan).

[1] Fermé C, Eghbali H, Meerwaldt JH, Rieux C, Bosq J, Berger F, et al. Chemotherapy plus involved‐field radiation in early‐stage Hodgkin’s disease. New England Journal of Medicine 2007;357:1916‐1927

Annunziata 2016

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective, single‐centre study

Study centre(s)

  • Not reported

Country

  • Italy

Median follow‐up time (range)

  • Not reported

Participants

Number of included participants

  • 68

Inclusion criteria

  • HL diagnosis

Exclusion criteria

  • Not reported

Consent

  • Not reported

Recruitment period

  • January 2007 to December 2014

Age (range, in years)

  • 39 (16‐72)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • ABVD according to the presence of risk factors defined by the European Organisation for Research and Treatment of Cancer (EORTC)

  • Favourable group (age < 50 years with ≤ 3 involved nodal areas, absence of mediastinal bulk (mediastinum‐to‐thorax ratio < 0.35), and erythrocyte sedimentation rate (ESR) < 50 mm without B symptoms or ESR < 30 mm with B symptoms): 3 cycles ABVD followed by radiotherapy, or 4 cycles ABVD without radiotherapy

  • Unfavourable group (age ≥ 50 years, > 4 involved nodal areas, presence of mediastinal bulk (mediastinum to‐thorax ratio ≥ 0.35), or ESR ≥ 50 mm without B symptoms or ESR ≥ 30 mm with B symptoms): 4 cycles ABVD followed by radiotherapy, or 6 cycles ABVD without radiotherapy

(therapy regimen reported in Raemaekers 20141)

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Half‐body PET scan (base of the skull to mid‐thigh)

Timing of prognostic factor measurement

  • Around day 25 (mean, range 22‐27) after cycle 1 of ABVD

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Scores of 1‐3 considered negative, scores of 4‐5 considered positive

  • 2 nuclear medicine physicians interpreted all scans

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), with progression during treatment, lack of complete remission at the end of first‐line treatment, and relapse counted as adverse events (AE)

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 2 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: all

Statistical method

  • Receiver operating characteristic (ROC) approach

  • Kaplan‐Meier (survival analysis)

  • Log‐rank (differences between groups)

  • Cox proportional hazards model

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Unclear

  • Clear description of participants and study characteristics. No inclusion and exclusion criteria provided.

Study attrition

  • Unclear risk

  • No loss to follow‐up reported. No length of follow‐up reported.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Stated in methods section that multiple factors were taken into account for analysis, but unclear which variables and how adjustment was conducted. Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • Poorly reported. Unclear whether multivariable analysis was reported.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • The authors declare that they have no conflict of interest.

Funding

  • Not reported

[1] Raemaekers JM, André MP, Federico M, Girinsky T, Oumedaly R, Brusamolino E, et al. Omitting radiotherapy in early positron emission tomography–negative stage I/II Hodgkin lymphoma is associated with an increased risk of early relapse: clinical results of the preplanned interim analysis of the randomised EORTC/LYSA/FIL H10 trial. Journal of Clinical Oncology 2014;32(12):1188‐1194

Barnes 2011

Study characteristics

Methods

Secondary citation(s)

  • Sher 2009

Language of publication

  • English

Study design

  • Retrospective, multi‐centre study (2 centres)

Study centre(s)

  • Massachusetts General Hospital Cancer Center and Dana‐Farber Cancer Institute, Massachusetts, USA

Country

  • USA

Median follow‐up time (range)

  • 46 months

Participants

Number of included participants

  • 96

Inclusion criteria

  • Diagnosed with classic, histology‐proven HL

  • Adults

  • Limited‐stage non‐bulky disease (mass < 10 cm)

  • ABVD chemotherapy

  • Availability of interim PET and end‐of‐treatment PET

Exclusion criteria

  • Nodular lymphocyte predominant HL

Consent

  • Not reported

Recruitment period

  • January 2000 to December 2008

Age (range, in years)

  • 34 (18‐77)

Ethnic group(s)

  • Not reported

Stages of disease

  • Early stages (I to IIB)

Comorbidities

  • Not reported

Therapy regimen

  • 4 or 6 cycles of ABVD with or without IFRT

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Whole‐body PET scan (base of the skull to mid‐thighs)

Timing of prognostic factor measurement

  • After 2 to 4 treatment cycles

Method for measurement (use of specific scale and cut‐off)

  • 2 nuclear medicine physicians interpreted all scans, final result based on consensus

  • Grading on a 4‐point scale with scores 0 or 1 considered negative and scores 2 to 4 considered positive

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Overall survival (OS), defined as the time from initial pathological diagnosis to death from any cause

  • Progression‐free survival (PFS), defined as time from diagnosis to progression or death from any cause

Secondary outcome(s) and definition(s)

  • Overall response rate (ORR), defined as number of subjects with either complete response (CR) or partial response (PR)

  • Primary refractory disease, defined as progressive disease on treatment or relapse within 3 months of completing therapy

Timing of outcome measurement

  • Unclear: 4 years reported in text, 10 years reported in figure

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • Not reported

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • OS: all

  • PFS: all

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank test

  • Fisher’s exact test (CR)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Unclear risk

  • Description of participants provided. Missing interim and end‐of‐treatment PET was part of the exclusion criteria. No comparison of baseline study sample (n = 155) and participants (n = 96) included. No reasons for missing scans provided.

Study attrition

  • Low risk

  • No loss to follow‐up.

Prognostic factor measurement

  • Moderate risk

  • Adequate measurement and description. No standardised criteria, but description of scoring system used. Prognostic factor measured the same way for all participants. Blinding not reported.

Outcome: Overall survival

Outcome measurement

  • High risk

  • Clear definition. Reporting of timing inconsistent (4 vs. 10 years).

'Other prognostic factors (covariates)'

  • Low risk

Statistical analysis and reporting

  • High risk

  • Statistical method appropriate for the data, but discrepancies between text and graphs detected.

Outcome: Progression‐free survival

Outcome measurement

  • High risk

  • Clear definition. Reporting of timing inconsistent (4 vs. 10 years).

'Other prognostic factors (covariates)'

  • Low risk

Statistical analysis and reporting

  • High risk

  • Statistical method appropriate for the data, but discrepancies between text and graphs detected.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • There are no relevant conflicts of interests to disclose.

Funding

  • Not reported

Casasnovas 2019

Study characteristics

Methods

Secondary citation(s)

  • Casasnovas 2018

Language of publication

  • English

Study design

  • Open‐label, randomised phase 3 trial

Study centre(s)

  • Multicentre (90 centres)

Countries

  • Belgium, France

Median follow‐up time (range)

  • 50.4 months (IQR: 42.9‐59.3) for all participants, not reported separately for standard treatment group

Participants

Number of included participants

  • 823 in total

  • 413 in standard treatment group

Inclusion criteria

  • Age 16‐60 years

  • Newly diagnosed HL

  • ECOG performance status score < 3

  • Minimum life expectancy of 3 months

  • Ann Arbor disease stage III, IV, or IIB with a mediastinum‐to‐thorax ratio of 0.33 or greater or extranodal localisation

  • No previous treatment for HL

  • Baseline PET (PET0) with at least one hypermetabolic lesion

  • Negative HIV, hepatitis C virus, and human T‐lymphotropic serology

  • Normal liver, renal, and haematological functions except for abnormalities related to HL

Exclusion criteria

  • Nodular lymphocyte predominant subtype

  • Severe cardiopulmonary or metabolic disease

Consent

  • Written, informed consent

Recruitment period

  • 19 May 2011 to 29 April, 2014

Age (range, in years)

  • 31 (IQR; ranges 23 ‐ 41)

Ethnic group(s)

  • Not reported

Stages of disease

  • II to IV, with B symptoms

Comorbidities

  • Not reported

Therapy regimen

  • Standard treatment group: 4 cycles of BEACOPPescalated, irrespective of PET2 result. After PET4: If PET4‐negative: 2 further cycles of BEACOPPescalated, if PET4‐positive: salvage therapy.

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Whole‐body PET scan (groin to head)

Timing of prognostic factor measurement

  • 2 to 4 weeks after completion of cycles 2 and 4 of chemotherapy

Method for measurement (use of specific scale and cut‐off)

  • Deauville criteria, with scores 1 to 3 considered negative, and scores 4 or 5 considered positive; Independent central review by 3 expert reviewers, final decision was based on at least two concordant responses

Was the same definition and method for measurement used in all participants?

  • Yes; participants were scanned on the same camera for all PET scans

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Presumably yes, but not explicitly mentioned

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from randomisation to first progression, relapse, or death from any cause or last follow‐up

Secondary outcome(s) and definition(s)

  • Safety, not defined

  • Overall response, not defined

  • Event‐free survival, defined as the time from randomisation to the first documented disease progression, relapse, start of a new anti‐lymphoma therapy, death from any cause, or last follow‐up

  • Disease‐free survival, defined as the time that complete response was recorded to the date of first documented disease progression, relapse or death related to lymphoma, toxicity from the study treatment (including treatment‐related secondary cancer), unknown cause or last follow‐up

  • Overall survival, defined as the time from randomisation to death from any cause or last follow‐up

Timing of outcome measurement

  • PFS: at 5 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • Yes; N = 11 stopped treatment before PET2, and further N = 14 stopped treatment before PET4

If yes, how were missing data handled?

  • All 413 participants included in ITT analysis, N = 412 included in safety analysis, N = 372 included in per‐protocol analysis

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS, OS: N = 413 in ITT analysis, N = 372 in per‐protocol analysis

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank test

  • Cox proportional hazard regression models

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • PFS: 759 (all participants that had reviewed PET2 and PET4 scans; not reported separately for standard group after PET2 without treatment modification)

  • OS: not reported

Statistical method

  • Cox proportional hazards regression model

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 8

List of all candidate covariates

  • PET assessment (PET2 and PET4)

  • Sex

  • Age

  • Eastern Cooperative Oncology Group score

  • B symptoms

  • Ann Arbor disease stage

  • Bulky disease

  • International Prognosis Score

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Adequate description of study population and recruitment. Detailed inclusion criteria.

Study attrition

  • Low risk

  • Reasons for loss to follow‐up provided for most participants with missing data.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

  • Only advanced stages included.

Statistical analysis and reporting

  • Low risk

  • Statistical methods appropriate and analysis fully reported.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome determined based on investigator assessment.

'Other prognostic factors (covariates)'

  • Low risk

  • Only advanced stages included. Multivariable analysis conducted.

Statistical analysis and reporting

  • Low risk

  • Statistical methods appropriate and analysis fully reported.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • R‐OC has received grants, personal fees and non‐financial support from Gilead, Roche, and Takeda, personal fees and non‐financial support from Bristol‐Myers Squibb, Celegne, and Merck Sharpe & Dohme, and personal fees from Abbvie. PB has received personal fees from Bristol‐Myers Squibb, Merck Sharpe & Dohme, and Takeda, grants from Takeda Millenium, and non‐financial support from Roche. AS has received personal fees from Takeda. EN‐V has received personal fees from Keocyt and Sanofi. FM has received personal fees from Celegne, Gilead, Janssen, and Roche/Genentech. RD has received personal fees from Bristol‐Myers Squibb, Celegne, Gilead, Janssen, Karyopharm, Roche, Sanofi, and Takeda. MM has received personal fees from Roche China. The other authors declare no competing interests.

Funding

  • Programme Hospitalier de Recherche Clinique

Cerci 2010

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Prospective, single‐centre study

Study centre(s)

  • São Paulo University Clinics Hospital, Brazil

Country

  • Brazil

Median follow‐up time (range)

  • 36 months (32‐40)

Participants

Number of included participants

  • 104

Inclusion criteria

  • Newly diagnosed, biopsy‐proven, classic HL

Exclusion criteria

  • Pregnancy

Consent

  • Yes; written

Recruitment period

  • August 2005 to December 2007

Age (range, in years)

  • 28 (13‐82)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • ABVD 4‐6 cycles (stage I and II), 6‐8 cycles (stage III), 8 cycles (stage IV)

  • Radiation therapy (stage I or II with no adverse risk factors and treated with 4 cycles ABVD; participants with bulky disease)

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Whole‐body PET scan

Timing of prognostic factor measurement

  • After 2 cycles of ABVD, as late as possible within the week before start of cycle 3

Method for measurement (use of specific scale and cut‐off)

  • No specific scale indicated

  • 2 board‐certified nuclear medicine physicians interpreted all scans

  • PET‐negative defined as no pathologic 18F‐FDG uptake at any site; PET‐positive defined as presence of focal 18F‐FDG uptake not attributed to physiologic biodistribution

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • 3‐year event‐free survival (EFS), defined as the time from diagnosis to treatment failure (incomplete response after first‐line treatment, progression during therapy, relapse or death) or last follow‐up

Secondary outcome(s) and definition(s)

  • 3‐year overall survival (OS)

Timing of outcome measurement

  • At 3 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • EFS: all

  • OS: all

Statistical method

  • Log‐rank (probability of treatment failure)

  • Kaplan‐Meier (survival curves)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics, consecutive sampling and no participants excluded based on interim‐PET availability.

Study attrition

  • Low risk

  • Loss to follow‐up reported.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • No definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Univariable analysis for multiple prognostic factors showed significance of factor of interest, but no multivariable analysis performed. Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • Statistical method in univariable analysis appropriate for the data, but no figures, only table with prognostic values, sensitivity and specificity. Discrepancies detected between text and graphs.

Outcome: Event‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Univariable analysis for multiple prognostic factors showed significance of factor of interest, but no multivariable analysis performed. Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • Statistical method in univariable analysis appropriate for the data, but no figures, only table with prognostic values, sensitivity and specificity. Discrepancies detected between text and graphs.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • Not reported

Funding

  • Not reported

Gallamini 2014

Study characteristics

Methods

Secondary citation(s)

  • Agostinelli 2016, Biggi 2013, Gallamini 2006, Gallamini 2007

Language of publication

  • English

Study design

  • Retrospective, international, multi‐centre study (17 centres)

Study centre(s)

  • 17 academic institutions worldwide

Countries

  • Various

Median follow‐up time (range)

  • 37 months (2‐110)

Participants

Number of included participants

  • 260

Inclusion criteria

  • HL participants with early stage unfavourable disease (IIA with adverse prognostic factors) or advanced stage disease (IIB – IVB)

  • Staging with PET‐CT at baseline and after 2 courses of ABVD

  • No change of treatment according to PET2

  • Minimum follow‐up of 1 year after completion of first treatment

Exclusion criteria

  • Missing CT data, baseline PET, interim PET, CT or PET slices; poor quality PET images; miscellaneous reasons (n=9)

Consent

  • No; due to retrospective study design

Recruitment period

  • January 2002 to December 2009

Age (range, in years)

  • 37.3 (14‐82)

Ethnic group(s)

  • Not reported

Stages of disease

  • Early stage unfavourable (IIA HL with adverse prognostic factors)

  • Advanced stages (IIB – IVB)

Comorbidities

  • Not reported

Therapy regimen

  • 4‐8 cycles ABVD with or without involved‐field radiotherapy or consolidation radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • A median of 12.3 days (range, 7‐22) after cycle 2 of ABVD

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • PET negative defined as scores 1‐3, PET positive defined as scores 4 or 5

  • 6 reviewers interpreted all scans independently

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes

Outcome(s)

Primary outcome(s) and definition(s)

  • Disease progression, defined as new disease within 6 months of first‐line treatment

  • Relapse, defined as disease occurring 6 months or longer after achieving complete remission

  • Progression‐free survival (PFS), defined as time from diagnosis to either disease progression or relapse, or to death as a result of any cause, whichever occurred first

  • Overall survival (OS), defined as the time from diagnosis to death from any cause

Secondary outcome(s) and definition(s)

  • Inter‐observer agreement using the 5‐PS for PET2 interpretation

Timing of outcome measurement

  • At 3 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Yes

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: 260

Statistical method

  • Kaplan Meier survival curves with Mantel‐Haenszel, log‐rank, Wilcoxon and Tarone‐Ware tests

  • Univariable regression analyses

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • PFS: 260

Statistical method

  • Cox proportional hazards regression model

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 9

List of all candidate covariates

  • Bulky disease

  • Lymphocyte

  • Albumin

  • White blood cells

  • IPS (0‐2 vs. ≥3)

  • Continued complete remission (CR) vs. no CR

  • Lactate dehydrogenase

  • Bone marrow involvement

  • PET2

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • Length of follow‐up reported.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Blinding not reported.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants. Blinding not reported.

'Other prognostic factors (covariates)'

  • Low risk

  • Only unfavourable and advances stages included.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants. Blinding not reported.

'Other prognostic factors (covariates)'

  • Low risk

  • Only unfavourable and advances stages included.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • Not reported

Funding

  • Reporting incomplete

  • The authors would like to thank: ... Keosys company for providing the Positoscope (R) network to distribute images to reviewers.

Gandikota 2015

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective study

Study centre(s)

  • Not reported

Country/Countries

  • Not reported

Median follow‐up time (range)

  • 46 months (24‐126)

Participants

Number of included participants

  • 78

Inclusion criteria

  • Biopsy‐proven, early‐stage (IA to IIB) classic HL of any subtype with or without bulky disease

  • Age > 18 years

  • Completion of planned ABVD and radiation therapy

  • At least 24 months of follow‐up or until proven relapse if earlier

Exclusion criteria

  • None

Consent

  • No

Recruitment period

  • January 2000 to December 2012

Age (range, in years)

  • 43 (median; 22‐86)

Ethnic group(s)

  • Not reported

Stages of disease

  • Early stages (IA to IIB)

Comorbidities

  • Not reported

Therapy regimen

  • ABVD (number of cycles based on risk factors and institutional guidelines) followed by involved‐field or extended‐field radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • PET‐CT scan (from base of the skull to upper thigh)

Timing of prognostic factor measurement

  • After ABVD cycle 2 to 4 or at the end of chemotherapy

Method for measurement (use of specific scale and cut‐off)

  • 5‐point scale

  • PET negative defined as a score ≤ 3

  • Staff physicians who were unaware of patient outcomes reviewed all scans

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes

Outcome(s)

Primary outcome(s) and definition(s)

  • Outcomes relevant to this review were not explored in the study

Secondary outcome(s) and definition(s)

  • Not applicable

Timing of outcome measurement

  • Not applicable

Was the same definition and method for measurement used in all participants?

  • Not applicable

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not applicable

Missing data

Participants with any missing value?

  • Yes: one patient without baseline PET due to pregnancy; one patient without detectable disease on the baseline scan (excision of single site disease)

If yes, how were missing data handled?

  • One patient without detectable disease on the baseline scan did not receive follow‐up PET since not considered necessary

Analysis

Univariable analysis: No

Multivariable analysis: No

Risk of bias (QUIPS)

No risk of bias assessment, since outcomes relevant to this review were not explored in the study.

Notes

Conflict of interest

  • The authors made no disclosure.

Funding

  • No specific funding was disclosed.

Hutchings 2005

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Not reported

Study centre(s)

  • Guy’s and St. Thomas’ Hospital, London

Country

  • UK

Median follow‐up time (range)

  • 40.2 months (6‐125)

Participants

Number of included participants

  • 85

Inclusion criteria

  • Histologically‐confirmed HL

  • Early interim FDG‐PET scans

Exclusion criteria

  • None

Consent

  • Not reported

Recruitment period

  • May 1993 to January 2004

Age (range, in years)

  • 36.7 (15‐73)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • According to departmental protocols: mainly ABVD, number of cycles not reported; additional radiotherapy depending on stage and site of HL

  • Alternative therapy for participants without satisfactory remission during initial chemotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Half‐body PET scan (mid‐brain to upper thigh)

Timing of prognostic factor measurement

  • After 2 or 3 cycles of chemotherapy, within the second week of the interval between cycles or as late as possible before administration of the next cycle

Method for measurement (use of specific scale and cut‐off)

  • No specific scale indicated

  • 2 experienced nuclear medicine physicians interpreted all scans, differences decided by consensus

  • PET‐negative defined as no evidence of disease; PET‐positive defined as increased uptake suspicious for malignant disease; Minimal residual uptake (MRU) defined as low‐grade uptake not likely to represent malignancy

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from diagnosis to first evidence of progression or relapse, or to disease‐related death

  • Overall survival (OS), defined as the time from diagnosis to death from any cause

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 2 and 5 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • NA

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: all

  • OS: not reported

Statistical method

  • Kaplan‐Meier (survival curves)

  • Log‐rank (differences between groups)

  • Proportional hazards Cox regression analysis

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • PFS: all

  • OS: none

Statistical method

  • Kaplan‐Meier (survival curves)

  • Log‐rank (differences between groups)

  • Proportional hazards Cox regression analysis

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 4

List of all candidate covariates

  • Early interim PET

  • Ann Arbor stage

  • PET‐MRU vs. PET‐negative

  • PET‐positive vs. PET‐negative

Risk of bias (QUIPS)

Study participation

  • Unclear risk

  • All eligible participants included. Clear description of participants and study characteristics. No inclusion / exclusion criteria provided. No comparison to baseline population, and no explanation of missing scans provided.

Study attrition

  • Moderate risk

  • Loss to follow‐up (8 participants), but reasons not provided.

Prognostic factor measurement

  • Low risk

  • Adequate description. PET results separated into negative, positive and low MRU, which sometimes was considered negative (clearly stated in these cases). No clear cut‐off in numbers.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants. Participants lost to follow‐up were still included in analysis.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method in univariable analysis appropriate for the data.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants. Participants lost to follow‐up were still included in analysis.

'Other prognostic factors (covariates)'

  • Low risk

  • Adjusted for disease stage.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • Not reported

Funding

  • Not reported

Hutchings 2006

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Prospective, multi‐centre study (4 centres)

Study centre(s)

  • Copenhagen University Hospital, Rigshospitalet, Herlev Hospital, Aarhus University Hospital

Country

  • Denmark

Median follow‐up time (range)

  • 22.8 months (6.1‐40.8)

Participants

Number of included participants

  • Total: 99

  • With Interim‐PET: 77

Inclusion criteria

  • Newly diagnosed HL

  • Adults (≥18 years of age)

Exclusion criteria

  • Diabetes mellitus

  • Pregnancy

Consent

  • Yes; written

Recruitment period

  • November 2001 to June 2004

Age (range, in years)

  • 36.2 (18.6 – 74.0)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • Various therapy regimens: ABVD (91%), ABV/MOPP (3%), ABVD/COPP (3%), BEACOPPesc. (3%), PVAG (1%)

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Half‐body PET scan (mid‐brain to upper thigh)

Timing of prognostic factor measurement

  • Within the last week before start of cycle 3 (PET2) and before cycle 5 (PET4)

Method for measurement (use of specific scale and cut‐off)

  • No specific scale indicated

  • 2 experienced nuclear medicine physicians interpreted all scans, differences in interpretation decided by consensus

  • Definitions for PET‐positive and PET–negative not reported

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes; nuclear medicine physicians were blinded from all clinical information except diagnosis

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from diagnosis to first evidence of progression or relapse, or to disease‐related death

  • Overall survival (OS), defined as the time from diagnosis to death from any cause

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 2 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Yes; clinicians were blinded from the results of PET

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: all

  • OS: not reported

Statistical method

  • Kaplan‐Meier (survival curves)

  • Log‐rank (differences between groups)

  • Proportional hazards Cox regression analysis

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • PFS: all

  • OS: not reported

Statistical method

  • Kaplan‐Meier (survival curves)

  • Log‐rank (differences between groups)

  • Proportional hazards Cox regression analysis

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 3

List of all candidate covariates

  • Interim PET

  • Clinical stage

  • Extranodal disease

Risk of bias (QUIPS)

Study participation

  • High risk

  • Significant number of participants without PET (n = 22 out of total n = 99). Imbalance between groups with or without PET scan regarding stage of disease.

Study attrition

  • Low risk

  • Lack of compliance in a small number of participants (n = 7 out of n = 99), but not in the subjects included in PET2 analysis.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Significant number of participants without PET (n = 22 out of n = 99).

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Clear definition of outcome. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition of outcome. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

  • Adjusted for disease stage.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • The authors have no financial interests in products studied in this work.

Funding

  • Not reported

Hutchings 2014

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Prospective, multi‐centre study

Study centre(s)

  • Not reported

Countries

  • USA, Italy, Poland, Denmark

Median follow‐up time (range)

  • 29 months

Participants

Number of included participants

  • 126*

*Potential overlap of Danish participants with those included in Hutchings 2006

Inclusion criteria

  • Newly diagnosed classic HL

Exclusion criteria

  • None

Consent

  • Yes; written

Recruitment period

  • Not reported

Age (range, in years)

  • 34.1 (median, 16.8‐76.7)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • Early‐stage disease: 2‐4 cycles ABVD followed by radiotherapy, or 6 cycles ABVD

  • Advanced‐stage disease: 6‐8 cycles ABVD with or without consolidation radiotherapy, with exceptions (5 Danish participants treated with BEACOPPesc)

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Whole‐body PET scan

Timing of prognostic factor measurement

  • Within the last 5 days of cycle 1 (PET1) and cycle 2 (PET2) (US and Italian participants had PET2 only if PET1 was positive)

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Scores of 1‐3 considered negative, scores of 4‐5 considered positive

  • Baseline interpretation by an expert with access to clinical information, second interpretation by an independent expert from another country blinded to clinical information

Was the same definition and method for measurement used in all participants?

  • No; not all participants received PET2

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes; experts in both stages blinded to clinical outcome, baseline experts also blinded to clinical information

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), not defined

  • Overall survival (OS), not defined

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 2 and 3 years

Was the same definition and method for measurement used in all participants?

  • Not reported; unclear due to multi‐national study design

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariate analysis for each outcome

  • PFS: all

  • OS: all

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank (differences between groups)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • PFS: all

  • OS: none

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank (differences between groups)

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 3

List of all candidate covariates

  • Interim PET (positive or negative)

  • Extranodal involvement

  • Disease stage (early or advanced stage)

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Description of participants and study characteristics given. No inclusion and exclusion criteria. Consecutive sampling and no exclusion based on interim PET availability. Detailed description of treatment regimen.

Study attrition

  • Low risk

  • No loss to follow‐up.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method in univariable analysis appropriate for the data.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • No definition of outcome. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method in univariable analysis appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • The author(s) indicated no potential conflicts of interest.

Funding

  • Not reported

Kobe 2018

Study characteristics

Methods

Secondary citation(s)

  • Borchmann 2017

Language of publication

  • English

Study design

  • Open‐label, international, randomised phase 3 trial

Study centre(s)

  • 301 hospitals and private practices in five European countries

Countries

  • Germany, Switzerland, Austria, the Netherlands, Czech Republic

Median follow‐up time (range)

  • Not reported for entire study population

Participants

Number of included participants

  • Total: 2101

  • Qualified for randomisation: 1945

Inclusion criteria

  • Histologically proven primary diagnosis of HL

  • Advanced stages: stage IIB with one or both of the risk factors large mediastinal mass and extranodal lesions, or stage III or IV

  • No previous treatment for HL

  • Age 18‐60 years at inclusion

  • Normal organ function, except for HL‐related impairments

  • Negative HIV test

  • Negative pregnancy test

  • Life expectancy > 3 months

Exclusion criteria

  • Incomplete diagnosis of the disease stage

  • Prior or concurrent disease that prevents treatment according to protocol

  • HL as part of a composite lymphoma

  • Prior chemotherapy or radiation

  • Malignant disease within the last 5 years (exceptions: basalioma, carcinoma in situ of the cervix uteri, completely resected melanoma TNMpT1)

  • Pregnancy, lactation

  • Eastern Cooperative Oncology Group (ECOG) performance status > 2

  • Long‐term ingestion of corticosteroids or antineoplastic drugs

  • Patient's lack of accountability, inability to appreciate the nature, meaning and consequences of the trial and to formulate his/her own wishes correspondingly

  • Noncompliance: refusal of blood products during treatment, epilepsy, drug dependency, change of residence to abroad, prior cerebral injury or similar circumstances that appear to make protocol treatment or long‐term follow‐up impossible

  • Antiepileptic treatment

  • General intolerance of any protocol medication

  • Unsafe contraceptive methods

  • Relationship of dependence or employer‐employee relationship to the sponsor or the investigator

  • Commitment to an institution on judicial or official order

  • Participation in another interventional trial that could interact with this trial

Consent

  • Yes; written, including consent to participate in the trial and to storage of data and tissue samples

Recruitment period

  • 14 May, 2008 to 18 July 2014

Age (range, in years)

  • Not reported for entire study population (Borchmann 2017)

Ethnic group(s)

  • Not reported

Stages of disease

  • Advanced stages: stage III‐IV, or stage II with B symptoms and one or both risk factors of large mediastinal mass

Comorbidities

  • None, due to exclusion criteria

Therapy regimen

  • 6 or 8 cycles of eBEACOPP (standard arm)

  • 4 cycles of eBEACOPP or 8 cycles of eBEACOPP with rituximab (experimental arm)

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • Between day 17 and day 21 of cycle 2 of chemotherapy

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • PET negative defined as scores 1 or 2, PET positive defined as scores 3 to 5

  • A multidisciplinary panel of experts centrally interpreted all scans

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • No; assessors who were masked to local findings, centrally reviewed PET‐2 and CT scans as well as x‐rays and clinical information (Borchmann 2017)

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from completion of staging until progression, relapse, or death from any cause

Secondary outcome(s) and definition(s)

  • Overall survival (OS), defined as time from completion of staging until death from any cause

Timing of outcome measurement

  • At 3 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • Participants with progressive disease, denoted by DS5 (Deauville score 5), were taken off protocol

  • 505 participants treated before the protocol amendment in June 2011 were excluded from survival analysis

If yes, how were missing data handled?

  • Participants with missing data were excluded from analysis

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • OS: 722

  • PFS: 722

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Cox regression analysis (hazard ratios)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • OS: 722

  • PFS: 722

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Cox regression analysis (hazard ratios)

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 9

List of all candidate covariates

  • Clinical stage

  • B symptoms

  • Large mediastinal mass

  • Extra‐nodal involvement

  • Involvement of 3 or more nodal areas

  • Elevated erythrocyte sedimentation rate

  • International Prognosis Score

  • HL subtype

  • PET positivity (DS4 vs. 1‐3)

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • Length of follow‐up reported. Exclusion of participants due to safety amendment during the study.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

  • Only advanced stages included.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

  • Only advanced stages included.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • We declare no competing interests.

Funding

  • The HD18 trial was funded by the Deutsche Krebshilfe (No. 107957 and 110617) and the Swiss State Secretariat for Education, Research and Innovation (SERI), and supported by Roche Pharma AG (No. ML‐21683).

Markova 2012

Study characteristics

Methods

Secondary citation(s)

  • Markova 2009

Language of publication

  • English

Study design

  • Retrospective, single‐centre study

Study centre(s)

  • Prague, institution not reported

Country

  • Czech Republic

Median follow‐up time (range)

  • 52 months

Participants

Number of included participants

  • 69

Inclusion criteria

  • Newly diagnosed, histologically proven HL

  • Clinical stage IIB with large mediastinal mass and/or extranodal disease, stage III or IV

  • Age 18‐60 years

Exclusion criteria

  • Presence of any concurrent disease precluding protocol treatment

  • Composite lymphoma

  • Previous malignancy

  • Previous chemo‐ or radiotherapy

  • Pregnancy or lactation

  • Diabetes mellitus and elevated fasting blood sugar level >130 mg/dl (exclusion from PET)

Consent

  • Not reported

Recruitment period

  • January 2004 to February 2008

Age (range, in years)

  • 30.7 (± 8.4)

Ethnic group(s)

  • Not reported

Stages of disease

  • IIB to IVB

Comorbidities

  • None, due to exclusion criteria

Therapy regimen

  • Treatment according to the HD15 trial of the German Hodgkin Study Group (GHSG) randomly assigned to either 8 cycles of BEACOPPescalated, 6 cycles of BEACOPPescalated or 8 cycles of time‐condensed BEACOPP14baseline

  • Local radiotherapy for participants with partial remission with residual mass ≥2.5cm and positive PET scan after chemotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • After cycle 4 of chemotherapy, as close as possible to cycle 5

Method for measurement (use of specific scale and cut‐off)

  • A local nuclear medicine physician interpreted all interim‐PET scans

  • PET‐positive defined as focal or diffuse uptake above background in a location incompatible with normal anatomy or physiology, without a specific standardised uptake cut‐off value; PET‐negative defined as no uptake, or increased uptake at the site of residual mass with an intensity lower or equal to the mediastinal blood pool

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from diagnosis to the first evidence of progression or relapse, or death from any cause

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • After cycle 4, 6/8 and 3 months after completion of chemotherapy

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: all

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank test (comparison between groups)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • All eligible participants included. Clear description of participants and study characteristics. Consecutive sampling. Inclusion and exclusion criteria provided.

Study attrition

  • Low risk

  • No loss to follow‐up.

Prognostic factor measurement

  • Moderate risk

  • Prognostic factor measured differently: PET4 scans reviewed locally (at the centre) by one physician, whereas PET6/8 assessment included central review.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

  • Only advanced stages included.

Statistical analysis and reporting

  • Low risk

  • Statistical method in univariable analysis appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • Not reported

Funding

  • Not reported

Mesguich 2016

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective, multi‐centre study (2 centres)

Study centre(s)

  • Haut‐Lévêque Hospital and Bergonié Institute, Bordeaux, France

Country

  • France

Median follow‐up time (range)

  • 58.9 months

Participants

Number of included participants

  • 76

Inclusion criteria

  • Biopsy‐proven, classic HL

  • Availability of baseline, interim and end‐of‐treatment PET‐CT

Exclusion criteria

  • Treatment with chemotherapy different than ABVD

  • Planned treatment modification following int‐PET results

  • End‐PET performance > 6 months after end of treatment

Consent

  • No; waived because of retrospective design

Recruitment period

  • December 2005 to April 2011

Age (range, in years)

  • 37 (median; 14‐67)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • Various therapy regimens: 3, 4, 6 or 8 cycles of ABVD with or without radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • After 2, 3 or 4 treatment cycles

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Consensual reading of two nuclear medicine physicians

  • Two cut‐offs for interim PET positivity tested and compared: either scores 4 to 5 considered PET positive, or scores 3 to 5 considered PET positive

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from diagnosis to either failure of first‐line treatment, relapse or death

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 5 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • NA

Analysis

Univariable analysis: Yes

Total number of participants included in univariate analysis for each outcome

  • PFS: all

Statistical method

  • Kaplan Meier analysis curve

  • Log‐rank test

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • PFS: all

Statistical method

  • Cox proportional hazard models

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 3

List of all candidate covariates

  • Interim PET

  • Disease stage*

  • Bulky disease*

*2 separate models, each adjusted for one of the 2 covariates other than interim PET

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • No loss to follow‐up.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants. Blinding not reported.

'Other prognostic factors (covariates)'

  • Low risk

  • Adjusted for disease stage.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • None declared.

Funding

  • No funding was sought or received for this study.

Oki 2014

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective, single‐centre study

Study centre(s)

  • MD Anderson Cancer Center, Houston, Texas, USA

Country

  • USA

Median follow‐up time (range)

  • 45 months

Participants

Number of included participants

  • Total: 325

  • 229 participants with PET2 analysed

  • 96 participants with PET3 excluded post‐hoc

Inclusion criteria

  • Classic HL

  • Treatment with ABVD

  • Availability of interim PET scan

Exclusion criteria

  • Additional treatment (e.g. with brentuximab vedotin or rituximab) except for radiotherapy

Consent

  • Not reported

Recruitment period

  • January 2001 to May 2011

Age (range, in years)

  • Group I (early‐stage non‐bulky): 32 (median, 18‐77)

  • Group II (stage II bulky): 36 (20‐60)

  • Group III (advanced stage IPS ≤ 2): 30 (19‐79)

  • Group IV (advanced stage IPS ≥ 3): 49 (19‐84)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • ABVD with or without radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • After 2 or 3 cycles of ABVD

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Scores of 1‐3 considered negative, scores of 4‐5 considered positive

  • Independent assessment by 3 nuclear medicine physicians

Was the same definition and method for measurement used in all participants?

  • No, 10 participants had only PET without CT scan

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from diagnosis to disease progression, relapse or death from any cause

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 3 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • No

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: all

Statistical method

  • Kaplan‐Meier survival curves with log‐rank test per subgroup

  • Univariable Cox proportional hazard models

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • No loss to follow‐up.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • Exclusion of participants with PET3 during analysis due to lack of prognostic value. Stratification according to disease stage resulted in small sample sizes per subgroup.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • No conflict of interest to disclose for the study.

Funding

  • Not reported

Okosun 2012

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective, multi‐centre study (6 centres)

Study centre(s)

  • 6 centres in London, UK

Country

  • UK

Median follow‐up time (range)

  • 27 months

Participants

Number of included participants

  • 23

Inclusion criteria

  • Newly diagnosed, histologically confirmed classic HL

  • Advanced stage

  • HIV positivity

Exclusion criteria

  • None

Consent

  • Not reported

Recruitment period

  • June 2007 to August 2010

Age (range, in years)

  • 42 (median, 32‐60)

Ethnic group(s)

  • Not reported

Stages of disease

  • Advanced stages: stage III –IV or stage IIB with at least one adverse prognostic factor

Comorbidities

  • HIV positive participants only

Therapy regimen

  • Treatment for HL: standard ABVD therapy

  • Treatment for HIV: HAART (two NRTIs in combination with either a non‐NRTI or a boosted protease inhibitor) antiretroviral therapy; G‐CSF per centre protocol; prophylaxis for Pneumocystis jiroveci

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Half‐body PET‐CT scan

Timing of prognostic factor measurement

  • After 2‐3 cycles of ABVD, within the week before start of the next cycle

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Scores 1‐3 considered negative, scores 4‐5 considered positive

  • Assessed at 3 established PET centres by own nuclear medicine physician and central review by nuclear medicine expert

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from diagnosis to disease progression or relapse or last follow‐up

Secondary outcome(s) and definition(s)

  • Overall survival (OS), defined as the time from diagnosis to death from any cause

  • Complete remission, defined as the disappearance of all disease manifestations at the end of therapy

Timing of outcome measurement

  • At 2 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariate analysis for each outcome

  • PFS: all

  • OS: not applicable, since no participants died

Statistical method

  • Kaplan‐Meier survival curves with log‐rank test

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics. Three participants did not have a staging PET, no reasons for missing PET provided.

Study attrition

  • Low risk

  • No loss to follow‐up. Length of follow‐up reported. Participants without interim PET (n = 11) excluded.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

  • Only unfavourable and advanced stages included.

Statistical analysis and reporting

  • High risk

  • Small sample size for some events (only two participants with positive interim PET result).

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • All authors have no conflicts of interest or disclaimers to declare.

Funding

  • Not reported

Orlacchio 2012

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective, single‐centre study

Study centre(s)

  • Policlinico Universitario TorVergata, Rome, Italy

Country

  • Italy

Median follow‐up time (range)

  • Not reported

Participants

Number of included participants

  • 132

Inclusion criteria

  • HL diagnosis based on biochemical tests and bone marrow biopsy

  • PET‐MDCT staging examination, interim PET‐MDCT and end of treatment PET‐MDCT performed

Exclusion criteria

  • None

Consent

  • Not reported

Recruitment period

  • January 2005 to June 2010

Age (range, in years)

  • 34 (mean, 16‐74)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • ABVD dose dependent on disease stage: stages I‐IIA 4x ABVD with radiotherapy; stages IIB‐IV 6‐8x ABVD with radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • PET scan from pelvis to head

Timing of prognostic factor measurement

  • At the end of the second ABVD cycle

Method for measurement (use of specific scale and cut‐off)

  • International Harmonization Project guidelines

  • Rated by a radiologist and nuclear medicine specialist, confirmation by semi‐quantitative analysis

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Complete remission, defined as the disappearance of symptoms and metabolic activity at any nodal or extranodal site with negative bone marrow biopsy

  • Partial remission, defined as persistence of significant metabolic activity at one site only, with at least 50% reduction in volume of the nodal masses or parenchymal nodular formations and persistence of disease at bone marrow level

  • Stable disease, defined as unchanged metabolic findings

  • Disease progression, defined as the appearance of new sites of pathological uptake and as a 50% increase in volume of nodal masses or previously detected parenchymal localisations

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At the end of treatment

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • NA

Analysis

Univariable analysis: Yes

Total number of participants included in univariate analysis for each outcome

  • Outcomes selected for univariable analysis unclear

Statistical method

  • Sensitivity, specificity, PPV, NPV

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

No risk of bias assessment, since outcomes relevant to this review were not explored in this study.

Notes

Conflict of interest

  • None

Funding

  • Not reported

Rossi 2014

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective, single‐centre study

Study centre(s)

  • Hospital of Dijon, France

Country

  • France

Median follow‐up time (range)

  • 50 months (22‐71)

Participants

Number of included participants

  • 59

Inclusion criteria

  • First diagnosis of classic HL

Exclusion criteria

  • Positive serology for HIV

Consent

  • Yes; written informed consent

Recruitment period

  • January 2007 to January 2010

Age (range, in years)

  • 35.5 (16‐76)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported, except for exclusion of HIV positive participants

Therapy regimen

  • Anthracycline‐based chemotherapy dependent on disease stage: stages I‐II 4‐6x chemotherapy with radiotherapy; stages III‐IV 8x chemotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Whole‐body PET‐CT scan

Timing of prognostic factor measurement

  • After 2 cycles of chemotherapy

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Scores 1‐3 considered negative, scores 4‐5 considered positive

  • ΔSUVmax (PET0‐PET2) dichotomized by applying the ROC approach

  • Independent review by 2 nuclear medicine physicians

Was the same definition and method for measurement used in all participants?

  • Different scanner used for 4 participants

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the time from the beginning of treatment until progression, relapse, or death from any cause or the date of last follow‐up

  • Time to progression (TTP), defined as time from the date of the first course of chemotherapy to any treatment failure, including progression, relapse, or death related to lymphoma, or the date of last follow‐up (participants with death from other cause were censored at the time of death)

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 4 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariate analysis for each outcome

  • PFS: all

Statistical method

  • Kaplan‐Meier product limit method with log‐rank test

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • PFS: all

Statistical method

  • Cox proportional hazards regression models per outcome

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 2

List of all candidate covariates

  • ΔSUVmax (PET0‐PET2)

  • International prognosis score (IPS)

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • No loss to follow‐up.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants. Blinding not reported.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • The costs of publication of this article were defrayed in part by the payment of page charges. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734. No potential conflict of interest relevant to this article was reported.

Funding

  • Not reported

Simon 2016

Study characteristics

Methods

Secondary citation(s)

  • Miltenyi 2015

Language of publication

  • English

Study design

  • Retrospective study

Study centre(s)

  • Not reported

Country

  • Hungary

Median follow‐up time (range)

  • 47.52 months (11‐80)

Participants

Number of included participants

  • 121

Inclusion criteria

  • Newly diagnosed HL

  • No previous treatment

Exclusion criteria

  • Immunosuppressive medications

  • Immunodeficiency

Consent

  • No

Recruitment period

  • 2007 to 2013

Age (range, in years)

  • 36.7 (mean, 17‐79)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • None due to exclusion criteria

Therapy regimen

  • ABVD dependent on disease stage: 6 or 8 cycles of ABVD, or 4 or 6 cycles of ABVD combined with radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • After cycle 2 of ABVD between days 11 and 14

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Scores 1‐3 considered negative, scores 4‐5 considered positive

  • Person(s) interpreting the scans not reported

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Overall survival (OS), not defined

  • Progression‐free survival (PFS), not defined

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 5 years after diagnosis

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • NA

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • OS: all

  • PFS: all

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank test (comparison between groups)

  • Cox proportional hazard model (effect of variants on survival)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: Yes

Total number of participants included in multivariable analysis for each outcome

  • OS: all

  • PFS: all

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank test (comparison between groups)

  • Cox proportional hazard model (effect of variants on survival

How was the prognostic factor treated?

  • Binary

Number of candidate covariates

  • 8

List of all candidate covariates

  • Age

  • Disease stage

  • Gender

  • B symptoms

  • Bulky disease

  • Treatment

  • PET2 positivity

  • Lymphocyte/monocyte ratio (LMR)

Risk of bias (QUIPS)

Study participation

  • Unclear risk

  • Description of participants provided, but no in‐ and exclusion criteria provided. Not clear how many participants were sampled and included from the baseline sample.

Study attrition

  • Low risk

  • No dropouts.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • Statistical analysis appropriate for the data. All primary outcomes reported, but discrepancies between text and graphs/tables detected.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • Statistical analysis appropriate for the data and all primary outcomes reported. However, discrepancies between text and graphs/tables detected.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • None of the authors have any competing interest in the manuscript.

Funding

  • Not reported

Straus 2011

Study characteristics

Methods

Secondary citation(s)

  • Kostakoglu 2012

Language of publication

  • English

Study design

  • Prospective phase 2, multi‐centre (29 centres), clinical trial

Study centre(s)

  • 29 Cancer and Leukemia Group B (CALGB) institutions

Country/Countries

  • Not reported

Median follow‐up time (range)

  • Not reported

Participants

Number of included participants

  • Total: 99

  • With interim‐PET: 88

Inclusion criteria

  • Previously untreated, histologically confirmed, classic HL with clinical stages I or II, measurable through physical examination or imaging studies

Exclusion criteria

  • Bulky disease

Consent

  • Yes; written

Recruitment period

  • 15 May 2004 to 29 September 2006

Age (range, in years)

  • 37 (18‐80)

Ethnic group(s)

  • Not reported

Stages of disease

  • Stages I ‐ IIB

Comorbidities

  • Not reported

Therapy regimen

  • 6 cycles of AVG administered on days 1 and 15 per cycle

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • 1 to 2 weeks after completion of cycle 2 of AVG

Method for measurement (use of specific scale and cut‐off)

  • Visual assessment was performed using International Harmonization Project criteria

  • Central review by 2 independent reviewers and an adjudicator

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes

Outcome(s)

Primary outcome(s) and definition(s)

  • Complete response, defined as complete remission or complete remission unconfirmed after 6 cycles of chemotherapy

Secondary outcome(s) and definition(s)

  • Progression‐free survival (PFS), measured from study entry until relapse

  • Adverse events (AEs), defined as toxicity including grade 3 or greater myelosuppression

Timing of outcome measurement

  • At 3 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • None

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • Complete response: none

  • PFS: 88

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank test (comparison between groups)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • Loss to follow‐up reported (n = 2).

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. PET2 available for n = 88 out of a total of n = 99 participants.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • Low risk

  • Only stages I ‐ IIB included.

Statistical analysis and reporting

  • Low risk

  • Statistical method in univariable analysis appropriate for the data.

Notes

Conflict of interest

  • The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked ‘‘advertisement’’ in accordance with 18 USC section 1734.The authors declare no competing financial interests.

Funding

  • This work was supported by the National Cancer Institute: CA77651 (D.J.S., H.S.), CA33601 (J.L.J.), CA32291 (A.S.L., G.P.C.), CA77440 (N.L.B.), CA04457 (L.K.), CA77658 (N.C.H., S.‐H.J.), CA32291 (R.W.T.), CA47642 (M.E.J.), and CA77597 (B.D.C.). This work was supported in part by the Lymphoma Foundation, Adam Spector Fund for Hodgkin Research, the Ernest & Jeanette Dicker Charitable Foundation, and Mr Daniel Moon and Family (for D.J.S.). This work was also supported by CALGB (National Cancer Institute) with partial support by Eli Lilly and Company. The research for CALGB 50203 was supported in part by grants from the National Cancer Institute (CA31946) to the CALGB (Dr Monica M. Bertagnolli, Chair) and to the CALGB Statistical Center (Dr Stephen George, CA33601). The content of this manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute.

Touati 2014

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Retrospective, single‐centre study

Study centre(s)

  • University Hospital of Limoges, France

Country

  • France

Median follow‐up time (range)

  • 65.8 months (2.2‐194.5)

Participants

Number of included participants

  • Total: 158

  • With interim‐PET: 68

Inclusion criteria

  • Histologically proven, classic HL

Exclusion criteria

  • Nodular lymphocyte predominant HL

Consent

  • Not reported

Recruitment period

  • February 1995 to July 2011

Age (range, in years)

  • 38 (16‐85)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • According to the standard of care at the time of diagnosis therapy regimens included ABVD, MOPP/ABV hybrid or BEACOPP; number of cycles not reported

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • After cycle 2 of chemotherapy

Method for measurement (use of specific scale and cut‐off)

  • Visual evaluation

  • PET‐positive if focal or diffuse accumulation of FDG in lesions higher than in surrounding tissue

  • FDG‐PET‐CT data (2005 and later) retrospectively reinterpreted using the Deauville 5‐point scoring system

Was the same definition and method for measurement used in all participants?

  • Different PET imaging techniques over time (dual‐head coincidence until 2005, then FDG‐PET‐CT), quality assurance and quality control program to ensure comparability of methods

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as time from date of diagnosis until relapse or death

  • Overall survival (OS), defined as time from first day of diagnosis until death from any cause

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 5 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: 68

  • OS: 68

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Chi‐squared test or t‐test (differences between groups)

  • ANOVA (comparison of means)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Unclear risk

  • Availability of interim PET as part of inclusion criteria, but not clear why less than 50% of participants had interim PET data. No comparison of baseline study sample (n = 357) with included participants (n = 158).

Study attrition

  • Low risk

  • All participants with available interim PET included.

Prognostic factor measurement

  • Moderate risk

  • Retrospective reinterpretation of PET scans using the Deauville criteria. Method described, but unclear whether assessors were blinded to initial interpretation.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • Not reported

Funding

  • This work was supported by the University Hospital of Limoges, CHU Limoges, F‐87042 France.

Ying 2014

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • Chinese, translated to English

Study design

  • Retrospective study

Study centre(s)

  • Peking University Cancer Hospital

Country

  • People’s Republic of China

Median follow‐up time (range)

  • 29.4 months (12.2‐52.4)*

*For the whole population (n = 50), but only 35 participants underwent interim PET

Participants

Number of included participants

  • Total: 50

  • With interim PET: 35

Inclusion criteria

  • Newly diagnosed HL according to the 2008 WHO Hematopoietic and Lymphoid Tissue Classification

Exclusion criteria

  • Not reported

Consent

  • Not reported

Recruitment period

  • September 2009 to December 2012

Age (range, in years)

  • 33 (14‐74)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • ABVD or BEACOPP with or without radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • From the top of the head to the middle thigh, the entire lower extremity was scanned if necessary

Timing of prognostic factor measurement

  • After 2 to 4 cycles of treatment

Method for measurement (use of specific scale and cut‐off)

  • Interpretation of scans by 2 experienced PET‐CT physicians

  • Scale and cut‐off not reported

Was the same definition and method for measurement used in all participants?

  • Not reported

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Not reported

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as the interval from diagnosis to first signs of tumour progression, patient death, or end of follow‐up

Secondary outcome(s) and definition(s)

  • None

Timing of outcome measurement

  • At 3 years

Was the same definition and method for measurement used in all participants?

  • Unclear, follow‐up was conducted via telephone and/or outpatient visits

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Not reported

Missing data

Participants with any missing value?

  • Only 35/50 participants underwent interim PET

If yes, how were missing data handled?

  • Not reported

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: 35

Statistical method

  • Kaplan‐Meier curves and life tables (survival analysis)

  • Log‐rank tests (comparison between groups)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • No loss to follow‐up. Length of follow‐up reported.

Prognostic factor measurement

  • Moderate risk

  • Adequate measurement and description, but no standardised criteria for PET scan evaluation.

Outcome: Overall survival

Not reported

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition. Outcome assessed differently for some participants (via telephone and/or outpatient visits).

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • Poor reporting of univariable analysis.

Outcome: Adverse events

Not reported

Notes

Translated from Chinese to English by Yu‐Tian Xiao.

Conflict of interest

  • Not reported

Funding

  • This study was funded by Natural Science Foundation of China (NSFC) grant no. 81470328 and Youth Fund of NSFC grant no. 81600162, 81600130.

Zaucha 2017

Study characteristics

Methods

Secondary citation(s)

  • NA

Language of publication

  • English

Study design

  • Prospective, observational, multi‐centre study (11 centres)

Study centre(s)

  • 11 haemato‐oncology centres

Country

  • Poland

Median follow‐up time (range)

  • 44.7 months (12.7–90.2)*

*Data for surviving participants only

Participants

Number of included participants

  • 310 registered participants, out of which 24 were excluded from analysis due to treatment intensification based on PET1 and/or clinical symptoms of active HL

Inclusion criteria

  • Newly diagnosed with classic HL

Exclusion criteria

  • Absent/poor‐quality PET‐CT images

Consent

  • Yes; written informed consent

Recruitment period

  • January 2008 to October 2014

Age (range, in years)

  • 30.8 (median, 18–80)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Not reported

Therapy regimen

  • ABVD dependent on disease stage: stages I‐IIA 2‐4x ABVD with radiotherapy or 6x ABVD; stages IIB‐IV 6‐8x ABVD with or without radiotherapy

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Whole‐body scan (mandibular angle to one third upper femur)

Timing of prognostic factor measurement

  • 11‐13 days after end of ABVD cycle 1 (PET1)

  • Additional scan after ABVD cycle 2 for participants with a PET1 score of 3‐5 (PET2)

Method for measurement (use of specific scale and cut‐off)

  • Deauville 5‐point scoring system

  • Scores 1‐3 considered negative, scores 4‐5 considered positive

  • 6 reviewers interpreted all scans using the blinded independent central review method, disagreements were resolved in a joint session

Was the same definition and method for measurement used in all participants?

  • No; PET2 only administered to participants with a PET1 score of 3‐5

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • Yes

Outcome(s)

Primary outcome(s) and definition(s)

  • Progression‐free survival (PFS), not defined

Secondary outcome(s) and definition(s)

  • Kinetics of response

Timing of outcome measurement

  • At 3 years

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • Yes

Missing data

Participants with any missing value?

  • Yes; only 198 participants had PET2 scans

If yes, how were missing data handled?

  • Not reported

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • 286 (PET1) / 198 (PET2)

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Cox proportional hazard regression analysis (HR between treatment groups)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

Prognostic factor measurement

  • Moderate risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants. However, while PET1 scans were available for all participants, the availability of PET2 scans was dependent on the result of PET1. No further scans were performed if PET1 was negative

Outcome: Overall survival

Not reported as a primary endpoint in the publication. IPD data were available and used to calculate the HR and SE for this outcome.

Outcome: Progression‐free survival

Outcome measurement

  • High risk

  • No definition of outcome.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • High risk

  • No detailed description of analysis.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • The authors have declared no conflicts of interest.

Funding

  • No funders to report.

Zinzani 2012

Study characteristics

Methods

Secondary citation(s)

  • Zinzani 2006

Language of publication

  • English

Study design

  • Retrospective, multi‐centre study (2 centres)

Study centre(s)

  • Bologna and Florence, Italy

Country

  • Italy

Median follow‐up time (range)

  • 45 months (6‐100)

Participants

Number of included participants

  • 304

Inclusion criteria

  • Diagnosed with HL

Exclusion criteria

  • Other treatment regimens than ABVD

  • Secondary lymphomas

  • Continuation of therapy during data analysis

Consent

  • Yes; written informed consent

Recruitment period

  • June 1997 to June 2009

Age (range, in years)

  • 32 (13‐78)

Ethnic group(s)

  • Not reported

Stages of disease

  • All stages

Comorbidities

  • Assessed, but not reported

Therapy regimen

  • ABVD dependent on disease stage: early stages 6x ABVD or 4x ABVD with radiotherapy; advanced stages 6x ABVD

Prognostic factor(s)

Prognostic factor(s)

  • Interim PET

Definition of prognostic factor(s)

  • Not reported

Timing of prognostic factor measurement

  • After cycle 2 of ABVD

Method for measurement (use of specific scale and cut‐off)

  • Juweid criteria

  • PET positive considered if focal FDG uptake that could not be attributed to physiological biodistribution, benign uptake or normal anatomy, with clearly increased activity relative to the background, excluding participants with minimal residual uptake

  • 2 experienced board‐certified nuclear medicine physicians interpreted all scans

Was the same definition and method for measurement used in all participants?

  • Yes

Were prognostic factor(s) assessed blinded for outcome(s), and for each other (if relevant)?

  • No

Outcome(s)

Primary outcome(s) and definition(s)

  • Response at the end of first‐line treatment and at follow‐up

Secondary outcome(s) and definition(s)

  • Progression‐free survival (PFS), defined as time from diagnosis to first observation of progressive disease or death from any cause

  • Overall survival (OS), defined as time from diagnosis to time of most recent visit or death

Timing of outcome measurement

  • At 9 years (for PFS and OS)

Was the same definition and method for measurement used in all participants?

  • Yes

Was/were outcome(s) assessed blinded for prognostic factor(s), and for each other (if relevant)?

  • No

Missing data

Participants with any missing value?

  • No

If yes, how were missing data handled?

  • Not applicable

Analysis

Univariable analysis: Yes

Total number of participants included in univariable analysis for each outcome

  • PFS: all

  • OS: all

Statistical method

  • Kaplan‐Meier (survival analysis)

  • Log‐rank test (comparison between groups)

How was the prognostic factor treated?

  • Binary

Multivariable analysis: No

Risk of bias (QUIPS)

Study participation

  • Low risk

  • Clear description of participants and study characteristics.

Study attrition

  • Low risk

  • No loss to follow‐up.

Prognostic factor measurement

  • Low risk

  • Adequate measurement and description. Prognostic factor measured the same way for all participants. No blinding of assessors.

Outcome: Overall survival

Outcome measurement

  • Low risk

  • Clear definition of outcome. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Progression‐free survival

Outcome measurement

  • Low risk

  • Clear definition of outcome. Outcome measured the same way for all participants.

'Other prognostic factors (covariates)'

  • High risk

  • Disease stage not accounted for.

Statistical analysis and reporting

  • Low risk

  • Statistical method appropriate for the data.

Outcome: Adverse events

Not reported

Notes

Conflict of interest

  • None

Funding

  • This work was partially supported by BolognAIL (Bologna, Italy).

ABVD: adriamycin/doxorubicin, bleomycin, vinblastine and dacarbazine; BEACOPP: bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine and prednisone; ePET: early positron emission tomography; FDG: [18F]‐fluorodeoxy‐D‐glucose; HL: Hodgkin lymphoma; HR: hazard ratio; IF‐RT: involved‐field radiation therapy; ITT: intention‐to‐treat; IQR: interquartile range; NPV: negative predictive value; OS: overall survival; PET: positron emission tomography; PET‐CT: positron emission tomography computed tomography; PFS: progression‐free survival; PPV: positive predictive value.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Adams 2016

Wrong publication type. Letter to the editor.

Adams 2017

Wrong publication type. Letter to the editor.

Adams 2018

Wrong publication type. Letter to the editor.

Adams 2018a

Wrong publication type. Letter to the editor.

Adams 2018b

Wrong publication type. Letter to the editor.

Adams 2019

Wrong publication type. Letter to the editor.

Advani 2007

Reported only end‐of‐chemotherapy PET scan results.

Afanasyev 2017

Wrong publication type. Protocol.

Albano 2017

PET‐adapted outcomes.

Albano 2018

PET‐adapted outcomes. Treatment was modified according to PET2 results.

Altamirano 2008

Wrong study population. Includes non‐Hodgkin lymphoma patients.

Ansell 2016

Wrong publication type. Article.

Awan 2013

Wrong patient population. Patients with any lymphoma were included; no separate data for Hodgkin lymphoma patients.

Bar‐Shalom 2003

Wrong study design. Comparison FDG PET and 67Ga scintigraphy.

Barrington 2011a

Wrong publication type. Meeting abstract.

Barrington 2017

Wrong publication type. Commentary.

Basu 2009

Wrong publication type. Commentary.

Becherer 2002

Wrong study design. End‐of‐chemotherapy PET. Includes non‐Hodgkin lymphoma.

Bednaruk‐Mlynski 2015

Wrong study design. Role of baseline PET/CT.

Biggi 2012

Wrong publication type. Conference abstract.

Biggi 2017

PET‐adapted outcomes. Treatment was modified according to PET2 results.

Bishop 2015

Wrong publication type. Commentary.

Bjurberg 2006

Wrong treatment. Retrospective study of patients with residual tumour or suspected relapse after therapy.

Blum 2002

Wrong patient population. Non‐Hodgkin lymphoma patients

Bodet‐Milin 2008

Wrong patient population. Non‐Hodgkin lymphoma patients.

Bodet‐Milin 2009

Wrong publication type. Article.

Boisson 2007

Wrong publication type. Article.

Borchmann 2016

Wrong study design. Literature review.

Bucerius 2006

Wrong publication type. Conference abstract.

Carras 2018

PET‐adapted outcomes. Treatment was modified according to PET2 results.

Ciammella 2016

PET‐adapted outcomes.

Cremerius 1999

Wrong study design. Retrospective study to validate the clinical value of FDG‐PET for therapy control.

Cremerius 2001

Wrong patient population. Patients with any lymphoma were included; no separate data for Hodgkin lymphoma patients.

Cuccaro 2016

PET‐adapted outcomes. Positive interim PET results led to change in therapy in three patients; data from these patients was not reported separately from the study population in analysis.

D'Urso 2018

Wrong study design. Analysis of metabolic parameters.

Damlaj 2017

PET‐adapted outcomes. Treatment was modified according to PET2 results.

Damlaj 2019

Pet‐adapted outcomes.

Danilov 2017

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Dann 2009

PET‐adapted outcomes.

Dann 2010

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Dann 2010a

PET‐adapted outcomes.Treatment was modified according to PET2 results.

Dann 2012

PET‐adapted outcomes.Treatment was modified according to PET2 results.

Dann 2013

PET‐adapted outcomes.Treatment was modified according to PET2 results.

Dann 2016

PET‐adapted outcomes. Treatment was modified according to PET2 results.

Dann 2017

PET‐adapted outcomes. Treatment was modified according to PET2 results.

Dann 2018

Wrong publication type. Response to letter.

deAndres‐Galiana 2015

Wrong study design. Prognostic factor identification study.

Diehl 2007

Wrong study design. The aim was to specify the negative predictive value of PET in patients with residual tumour mass after chemotherapy.

El‐Galaly 2012

Wrong study design. The study evaluated the utility of PET scans for post‐therapy routine surveillance imaging.

Evens 2014

Wrong publication type. Article.

Fanti 2008

Wrong study design. Case study.

Filmont 2003

Wrong patient population. Patients with aggressive lymphoma undergoing salvage therapy.

Fornecker 2017

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Freudenberg 2004

Wrong patient population. Patients with any lymphoma were included; no separate data for Hodgkin lymphoma patients.

Friedberg 2002

Wrong study design. The study intended to compare FDG‐PET to gallium scintigraphy in the staging and follow‐up of newly diagnosed patients with Hodgkin lymphoma.

Friedberg 2004

Wrong study design. The study intended to compare FDG‐PET to gallium scintigraphy in the staging and follow‐up of newly diagnosed patients with Hodgkin lymphoma.

Front 1999

Wrong treatment. The study investigated the utility of gallium scintigraphy performed early during treatment as a means to predict outcome and optimise treatment in Hodgkin lymphoma patients.

Fruchart 2006

Wrong patient population. Patients with B‐cell lymphoma.

Gallamini 2008

Wrong publication type. Article.

Gallamini 2017

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Gallamini 2018

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Gallamini 2018a

Wrong publication type. Reply to letter.

Gallowitsch 2008

Wrong publication type. Commentary.

Goldschmidt 2011

Wrong patient population. Relapsed, aggressive non‐Hodgkin lymphoma.

Greil 2018

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Guidez 2016

Wrong publication type. No abstract or full text.

Hagtvedt 2015

Wrong study design. Comparison between FDG‐PET and diffusion‐weighted magnetic resonance imaging for assessment of early treatment response in lymphoma.

Haioun 2005

Wrong patient population. Patients with any lymphoma were included; no separate data for Hodgkin lymphoma patients.

Hartmann 2012

Wrong study design. The study investigated protein expression patterns in different Hodgkin lymphoma subtypes.

Hartridge‐Lambert 2013

Wrong study design. The study evaluated the risk of disease recurrence and the value of radiologic surveillance in patients treated with ABVD alone who achieved a complete remission according to post‐treatment PET. PET was not treated as a prognostic factor.

Honda 2014

Wrong patient population. Letter to the editor, presenting the case of one patient with pulmonary Hodgkin lymphoma.

Hueltenschmidt 2001

Baseline and end‐of‐chemotherapy PET results.

Huic 2006

Wrong treatment. Patients within three months after completion of conventional initial therapy or salvage therapy with high‐dose chemotherapy were included in the study population; no subgroup analysis was reported.

Hutchings 2007

End‐of‐chemotherapy PET.

Iagaru 2008

Wrong patient population. Patients with any lymphoma were included; no separate data for Hodgkin lymphoma patients.

Illidge 2015

PET‐adapted outcomes. Commentary on a research news article about PET‐adapted treatment in Hodgkin lymphoma patients.

Jerusalem 2003

End‐of‐chemotherapy PET.

Johnson 2015

PET‐adapted outcomes.

Johnson 2016

PET‐adapted outcomes.

Kamran 2016

PET‐adapted outcomes.

Kamran 2018

PET‐adapted outcomes. Treatment was modified according to PET2 results. Data was not reported separately for PET‐positive and PET‐negative patients.

Kobe 2008

Wrong study design. The study evaluated the negative predictive value of PET scans in advanced‐stage Hodgkin lymphoma patients.

Kobe 2014

Wrong study design. The study evaluated how computed tomography might help improve the positive predictive value of PET in identifying potential high‐risk patients.

Kostakoglu 2006

Wrong patient population. Patients with either diffuse large cell lymphoma or Hodgkin lymphoma were included; no separate data for Hodgkin lymphoma patients.

Li 2013

Wrong patient population. The study population consisted of patients with mature T‐cell and natural killer cell lymphomas.

Lowe 2002

Wrong study design. Commentary.

Milgrom 2017

Wrong study design. The study population consisted mostly of PET‐positive patients. The study compared data from PET‐positive patients who received salvage chemotherapy or autologous stem cell transplantation with patients who received radiotherapy only.

Mocikova 2010

Wrong study design. The study evaluated the routine use of PET scans in Hodgkin lymphoma patients during follow‐up and in cases of suspected relapse.

Mocikova 2011

Wrong treatment. The study evaluated the prognostic significance of pre‐transplant PET scans after salvage chemotherapy before autologous stem cell transplant in patients with relapsed or refractory Hodgkin lymphoma.

Molnar 2010

End‐of‐chemotherapy PET.

Moskowitz 2015

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Naumann 2001

End‐of‐chemotherapy PET.

NCT00784537

PET‐adapted outcomes. Treatment was modified according to interim PET results.

NCT00795613

PET‐adapted outcomes. Treatment was modified according to interim PET results.

NCT01358747

PET‐adapted outcomes. Treatment was modified according to interim PET results.

NCT01652261

PET‐adapted outcomes. Study closed due to lack of recruitment.

NCT02292979

Wrong study design.

Nguyen 2017

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Panizo 2004

End of chemotherapy PET.

Paolini 2007

PET‐adapted outcomes.

Pavlovsky 2019

PET‐adapted outcomes.

Pichler 2000

Wrong study design. Comparison of FDG‐Hybrid‐PET scans.

Reinhardt 2005

Wrong study design. The study evaluated the accuracy of computed tomography and FDG‐PET for prediction of progression‐free survival of Hodgkin lymphoma and non‐Hodgkin lymphoma patients after completion of therapy.

Rigacci 2002

Wrong study design. Letter.

Rigacci 2017

Wrong study design. Letter.

Rubello 2015

Wrong study design. The study evaluated the variability of FDG liver uptake in patients with Hodgkin lymphoma.

Sakr 2017

Wrong study design.

Schot 2007

Wrong treatment. The study population included patients with recurring lymphoma who were treated with second‐line chemotherapy followed by autologous stem cell transplantation.

Simontacchi 2015

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Slaby 2002

Wrong patient population. Patients with any lymphoma were included; no separate data for Hodgkin lymphoma patients.

Spaepen 2001

Reported only end‐of‐chemotherapy PET scan results.

Specht 2007

Wrong publication type. Article.

Spinner 2018

Wrong publication type. Article.

Straus 2018

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Strigari 2016

Wrong study design. The aim of the study was to present a novel quantitative tool to refine the risk‐class assessment of the Deauville criteria.

Sucak 2011

Wrong treatment. The study population included patients with relapsed or refractory lymphoma post‐autologous stem cell transplantation.

Tirelli 2015

Wrong publication type. Article.

Tomita 2015

Wrong patient population. The study population consisted of patients with peripheral T cell lymphoma.

Torizuka 2004

PET‐adapted outcomes. Includes non‐Hodgkin lymphoma patients.

Trotman 2017

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Tseng 2012

Wrong patient population. The study population included relapsed patients.

Villa 2018

PET‐adapted outcomes. Treatment was modified according to interim PET results.

Weidmann 1999

Wrong patient population. Includes relapsed patients.

Wilson 2018

Wrong publication type. Commentary.

Xie 2018

Wrong publication type. Review.

Yasgur 2015

Wrong publication type. Commentary.

Yoshimi 2008

Wrong treatment. The study population included lymphoma patients with a poor prognosis who had received FDG‐PET scans within one month before allogeneic stem cell transplantation.

Zabrocka 2016

Wrong study design. The study evaluated the current usage of PET scans and its clinical usefulness at different points in Hodgkin lymphoma management based on a single‐institution experience.

Zaucha 2009

Wrong publication type. Review.

Zinzani 1999

Wrong patient population. Includes non‐Hodgkin lymphoma patients.

Zinzani 2002

Wrong patient population. Includes non‐Hodgkin lymphoma patients.

Zinzani 2016

PET‐adapted outcomes. Treatment was modified according to interim PET results.

ABVD: adriamycin/doxorubicin, bleomycin, vinblastine and dacarbazine; FDG: [18F]‐fluorodeoxy‐D‐glucose; PET: positron emission tomography; PET‐CT: positron emission tomography computed tomography.

Characteristics of studies awaiting classification [ordered by study ID]

Abramson 2010

Notes

Title: End of treatment but not interim PET scan predicts outcome in non‐bulky limited stage Hodgkin lymphoma. (Conference abstract)

Aim

  • To establish the prognostic value of interim PET scans in limited stage patients with non‐bulky disease

Study design

  • Retrospective

Country/treatment centre(s)

  • USA (Massachusetts General Hospital, Dana‐Farber Cancer Institute, Harvard School of Public Health, Boston MA)

Number of included participants

  • 96

Inclusion criteria

  • Non‐bulky limited stage cHL treated at the institutions between 2000 and 2008; Bulk was defined as a mass >=10 cm or >=1/3 of the intrathoracic diameter.

Exclusion criteria

  • None

Treatment

  • 4 to 6 cycles of ABVD with or without IFRT

Primary outcome measure(s)

  • Overall survival

  • Progression‐free survival

Algrin 2010

Notes

Title: Interim‐positron emission tomography with [18F]fluorodeoxyglucose (interim‐PET) evaluation in mediastinal lymphoma including Hodgkin lymphoma (HL) and primary mediastinal large B‐cell lymphoma (PMBL).

(Conference abstract)

Aim

  • To investigate the prognostic value of qualitative and semi‐quantitative evaluations of interim‐PET in mediastinal lymphoma

Study design

  • Retrospective

Country/treatment centre(s)

  • Not reported

Number of included participants

  • 48

Inclusion criteria

  • Previously untreated, age under 60 at diagnosis and at least one interim‐PET evaluation available

Exclusion criteria

  • Individuals with sub‐diaphragmatic or medullar localisations of lymphoma

Treatment

  • Not reported

Primary outcome measure(s)

  • Event‐free survival

Arce‐Calisaya 2013

Notes

Title: Interim FDG PET‐CT in Hodgkin's lymphoma ‐ Does binary response assessment criteria have any prognostic value?

(Conference abstract)

Aim

  • To evaluate whether binary response assessment criteria (positive or negative) has any prognostic significance after 2 cycles of ABVD therapy

Study design

  • Retrospective

Country/treatment centre(s)

  • UK

Number of included participants

  • 99

Inclusion criteria

  • Newly diagnosed adults with advanced‐stage HL undergoing baseline and interim (post‐2 cycles ABVD) 18F‐FDG PET‐CT

Exclusion criteria

  • None

Treatment

  • ABVD

Primary outcome measure(s)

  • Recurrence‐free survival after 1 year

Baratto 2015

Notes

Title: Interim‐PET in Hodgkin lymphoma: Deauville criteria and metabolic parameters as prognostic factors.

(Conference abstract)

Aim

  • To explore the prognostic role of i‐PET in individuals with HL

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy

Number of included participants

  • 83

Inclusion criteria

  • Newly diagnosed HL, stage I‐IV disease

Exclusion criteria

  • None

Treatment

  • Not reported

Primary outcome measure(s)

  • Overall survival

  • Disease‐free survival

Barna 2011

Notes

Title: Prognostic value of interim 18FDG‐PET‐CT in patients with Hodgkin's lymphoma using different 5‐point visual scales for interpretation.

(Conference abstract)

Aim

  • To compare the effect on prognosis of the currently applied MRU definitions

Study design

  • Prospective

Country/treatment centre(s)

  • Hungary

Number of included participants

  • 82

Inclusion criteria

  • Newly‐diagnosed HL

Exclusion criteria

  • None

Treatment

  • 6 courses of ABVB/EBVD, additional radiotherapy according to the protocol

Primary outcome measure(s)

  • Overall survival

  • Progression‐free survival

Barrington 2011

Notes

Title: Are the Deauville criteria a reliable tool for assessment of interim PET in Hodgkin lymphoma?

(Conference abstract)

Aim

  • To measure agreement between experienced reporters reading interim PET‐CT scans from an international cohort of patients according to the Deauville criteria

  • To measure progression‐free survival in advanced HL according to interim PET

Study design

  • Not reported

Country/treatment centre(s)

  • International study

Number of included participants

  • 262

Inclusion criteria

  • Individuals diagnosed with stage IIB‐IV HL

Exclusion criteria

  • None

Treatment

  • ABVD

Primary outcome measure(s)

  • Progression‐free survival

Bentur 2017

Notes

Title: The predictive value of interim PET‐CT in elderly patients with Hodgkin lymphoma.

This is an abstract only and a lot of relevant information is missing. A full‐text has not been published yet. It is particularly unclear whether participants have received treatment adaptation based on the interim PET result. Authors need to be contacted for more information.

Aim

  • To evaluate the significance of iPET in elderly individuals with HL

Study design

  • Retrospective study (1998 to 2016)

Country/treatment centre(s)

  • Unclear, multicentre study (5 centres)

Number of included participants

  • 95

Inclusion criteria

  • Individuals diagnosed with HL between 1998 to 2016

  • Older adults (>= 60 years)

Exclusion criteria

  • Not reported

Treatment

  • Fifty‐nine participants received first‐line treatment with ABVD, in 13 participants chemotherapy was followed by IVRT (treatment unclear for the remaining participants)

Primary outcome measure(s)

  • Overall survival

  • Progression‐free survival

  • Time frame: five years

Berenger 2010

Notes

Title: Prognostic value of interim 18F‐FDG PET‐CT in mediastinal bulky Hodgkin lymphoma.

(Conference abstract)

Aim

  • To determine if Negative Predictive Value (NPV) remains high in individuals who present with mediastinal bulky disease

Study design

  • Retrospective

Country/treatment centre(s)

  • France

Number of included participants

  • 38

Inclusion criteria

  • Previously untreated individuals with HL, with localiSed mediastinal bulky disease

Exclusion criteria

  • None

Treatment

  • Chemotherapy with or without additional radiotherapy

Primary outcome measure(s)

  • Progression‐free survival

  • NPV and PPV of iPET

Bhatwadekar 2017

Notes

Title: Excellent outcome in Hodgkin lymphoma with ABVD and CMT: A single‐centre retrospective analysis.

(Conference abstract)

Aim

  • To evaluate the outcome of individuals with HL receiving ABVD alone or in combination with RT

Study design

  • Retrospective

Country/treatment centre(s)

  • India (Haemato Oncology Care Centre, Vadodara)

Number of included participants

  • 63

Inclusion criteria

  • Not reported

Exclusion criteria

  • Not reported

Treatment

  • ABVD alone or in combination with RT

Primary outcome measure(s)

  • Overall survival

  • Progression‐free survival

Cimino 2014

Notes

Title: The complementary prognostic role of baseline and interim PET in predicting treatment outcome in advanced‐stage Hodgkin lymphoma.

(Conference abstract)

Aim

  • To evaluate the contribution of PET combined with computed tomography (PET‐CT) and contrast enhanced computed tomography (ceCT) in the staging and in the prognostication of untreated advanced HL

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy, Poland, Denmark (multicentre)

Number of included participants

  • 162

Inclusion criteria

  • Not reported

Exclusion criteria

  • Not reported

Treatment

  • ABVD with or without RT

Primary outcome measure(s)

  • Overall survival

  • Event‐free survival

Cocorocchio 2009

Notes

Title: Prognostic role of interim 18FDG‐PET in Hodgkin lymphoma: A single‐center experience.

(Conference abstract)

Aim

  • Single‐centre experience with using 18FDG‐PET as a prognostic factor for long term complete remission (CR)

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy

Number of included participants

  • 65

Inclusion criteria

  • Newly diagnosed with HL

Exclusion criteria

  • Not reported

Treatment

  • VBM or ChlVPP/ABVVP followed by IFRT

Primary outcome measure(s)

  • Complete remission

  • Freedom from treatment failure

Cocorocchio 2011

Notes

Title: Evaluation of interim 18FDG‐PET in advanced Hodgkin lymphoma (HL) patients (PTS) treated with ChlVPP/ABVVP regimen.

(Conference abstract)

Aim

  • To evaluate the prognostic value of interim 18 FDG‐PET in advanced HL patients treated with intensified ChlVPP/ABVVP

Study design

  • Not reported

Country/treatment centre(s)

  • Italy

Number of included participants

  • 70

Inclusion criteria

  • Not reported

Exclusion criteria

  • Not reported

Treatment

  • 6 cycles of ChlVPP/ABVVP

Primary outcome measure(s)

  • Overall survival

  • Freedom from treatment failure

Copeland 2010

Notes

Title: Single institution experience with interim PET evaluation in newly diagnosed CHL receiving ABVD chemotherapy: Need for standardization.

(Conference abstract)

Aim

  • To evaluate the use of interim PET for the identification of individuals with classic HL, who are at risk for relapse after first‐line therapy

Study design

  • Retrospective

Country/treatment centre(s)

  • USA (MD Anderson Cancer Center, Houston TX)

Number of included participants

  • 57

Inclusion criteria

  • Newly diagnosed cHL

Exclusion criteria

  • Not reported

Treatment

  • ABVD

Primary outcome measure(s)

  • Event‐free survival

Cuzzocrea 2015

Notes

Title: The Deauville criteria and metabolic parameters as prognostic factors in interim PET in Hodgkin lymphoma: A single centre experience.

(Conference abstract)

Aim

  • To explore the prognostic role of i‐PET in individuals with HL

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy

Number of included participants

  • 83

Inclusion criteria

  • Newly diagnosed HL, stage I‐IV disease

Exclusion criteria

  • Not reported

Treatment

  • Not reported

Primary outcome measure(s)

  • Overall survival

  • Disease‐free survival

De Rueda 2013

Notes

Title: Prognostic value of 18F‐FDG PET‐CT in Hodgkin lymphoma.

(Conference abstract)

Aim

  • To determine the value of 18F‐FDG PET‐CT after the second and sixth cycle of first line therapy with ABVD or BEACOPP in the outcome of individuals with HL

Study design

  • Retrospective, January 2007 to December 2012

Country/treatment centre(s)

  • Spain

Number of included participants

  • 79

Inclusion criteria

  • HL diagnosis

Exclusion criteria

  • Not reported

Treatment

  • ABVD or BEACOPP

Primary outcome measure(s)

  • Progression‐free survival

Fabbri 2011

Notes

Title: 'Early FDG‐PET' predicts clinical course of Hodgkin's lymphoma although does not correlate with macrophages infiltration in diagnostic specimens.

(Conference abstract)

Aim

  • To verify the prognostic role both of "early‐FDG PET" and of macrophagic infiltration, and to test if "early‐FDG PET" positivity could correlate with high macrophagic infiltration in diagnostic specimens

Study design

  • Retrospective, February 2007 to July 2010

Country/treatment centre(s)

  • Italy (Siena and Florence haematology departments)

Number of included participants

  • 52

Inclusion criteria

  • Diagnosed HL

Exclusion criteria

  • Not reported

Treatment

  • 4 to 6 cycles of ABVD with or without IFRT

Primary outcome measure(s)

  • Complete remission

  • CD68 expression

Fiore 2010

Notes

Title: Early interim FDG‐PET during intensified BEACOPP therapy for advanced‐stage Hodgkin disease shows a lower positive predictive value than during ABVD.

(Conference abstract)

Aim

  • To examine the predictive role on treatment outcome of early interim FDG‐PET in individuals with HL, treated with BEACOPP (4 escalated + 4 baseline cycles)

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy (8 haematological institutions)

Number of included participants

  • 44

Inclusion criteria

  • Diagnosed HL, advanced stage (IIB to IVB, or IIA with adverse prognostic factors)

Exclusion criteria

  • Not reported

Treatment

  • BEACOPP

Primary outcome measure(s)

  • Complete remission

  • Failure‐free survival

Gallegos 2012

Notes

Title: The importance of PET‐CT as method of evaluation of early response to treatment in HL.

(Conference abstract)

Aim

  • To assess the importance of PET‐CT as method of evaluation of early response to treatment in HL

Study design

  • Retrospective, 2002 to 2011

Country/treatment centre(s)

  • Spain (The Miguel Servet's Hospital, Zaragoza)

Number of included participants

  • 61

Inclusion criteria

  • Diagnosed HL, first‐line therapy

Exclusion criteria

  • Not reported

Treatment

  • ABVD or BEACOPP

Primary outcome measure(s)

  • Progression‐free survival

Hohaus 2015

Notes

Title: The risk of progression of Hodgkin lymphoma in patients with negative interim PET: A role for the number of tumor‐infiltrating macrophages (CD68+ cell counts) and B symptoms.

(Conference abstract)

Aim

  • To evaluate if integration of the response evaluation with iPET with parameters available at diagnosis could add prognostic information, allowing a better risk‐stratification of individuals with HL

Study design

  • Retrospective, 2007 to 2014

Country/treatment centre(s)

  • Italy (Università Cattolica del Sacro Cuore, Rome)

Number of included participants

  • 102

Inclusion criteria

  • Diagnosed classic HL

Exclusion criteria

  • Not reported

Treatment

  • ABVD

Primary outcome measure(s)

  • Progression‐free survival

Hutchings 2010

Notes

Title: correlation of FDG‐PET results after one cycle and after two cycles of chemotherapy in Hodgkin lymphoma.

(Conference abstract)

Aim

  • To study the correlation of PET results after one cycle and after two cycles of chemotherapy, and to investigate if the high predictive value of PET after two cycles is obtainable already after one cycle of chemotherapy

Study design

  • Prospective trial

Country/treatment centre(s)

  • Denmark (Copenhagen), USA (New York)

Number of included participants

  • 36

Inclusion criteria

  • Diagnosed HL

Exclusion criteria

  • Not reported

Treatment

  • ABVD or BEACOPPesc

Primary outcome measure(s)

  • Negative predictive value

Knight‐Greenfield 2013

Notes

Title: Interim FDG PET‐CT to predict progression‐free survival (PFS) better than clinical and baseline metabolic measurements in Hodgkin lymphoma (cHL).

(Conference abstract)

Aim

  • To determine the best predictor of PFS among various variables of tumour metabolic measurements at baseline and at interim PET‐CT compared to conventional methods in individuals with classic HL

Study design

  • Retrospective

Country/treatment centre(s)

  • Not reported

Number of included participants

  • 58

Inclusion criteria

  • Diagnosed classic HL, ABVD therapy, minimal follow‐up of 2 years

Exclusion criteria

  • Not reported

Treatment

  • ABVD

Primary outcome measure(s)

  • Progression‐free survival

Leontjeva 2016

Notes

Title: Significance of early interim PET results in advanced Hodgkin lymphoma treated intensive program EACOPP‐14.

(Conference abstract)

Aim

  • To evaluate the use of interim PET to guide treatment in advanced stage individuals with classic HL

Study design

  • Not reported, December 2009 to December 2013

Country/treatment centre(s)

  • Russia

Number of included participants

  • 36

Inclusion criteria

  • Newly diagnosed classic HL (stages IIB to IV, or IIA with bulk), adults

Exclusion criteria

  • Not reported

Treatment

  • EACOPP‐14 with or without RT

Primary outcome measure(s)

  • Progression‐free survival

Luminari 2010

Notes

Title: The use of FDG positron emission tomography (FDG‐PET) in patients with Hodgkin lymphoma (HL) in the "real world": A population based study from northern Italy.

(Conference abstract)

Aim

  • To assess how FDG‐PET is currently used in individuals with HL

Study design

  • Unclear, 2006 to 2008

Country/treatment centre(s)

  • Italy (Cancer Registries in Modena, Ferrara, Parma and Reggio Emilia)

Number of included participants

  • 136

Inclusion criteria

  • Diagnosed HL, adults (18 to 75 years), HIV negative

Exclusion criteria

  • None

Treatment

  • Not reported

Primary outcome measure(s)

  • Overall survival

  • Relapse‐free survival

  • Failure‐free survival

Luminari 2011

Notes

Title: Use of 2‐[18F]fluoro‐2‐deoxy‐D‐glucose positron emission tomography in patients with Hodgkin lymphoma in daily practice: a population‐based study from Northern Italy

Authors need to be contacted to clarify whether the treatment has been adapted based on the interim PET results.

Aim

  • To investigate how PET is currently used in daily practice and whether results obtained in clinical trials and retrospective series can be generalised to all individuals with HL.

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy

  • Participants were identified from archives of four population‐based Italian cancer registries

Number of included participants

  • Total: 136

Inclusion criteria

  • Registration in one of the four population‐based Italian cancer registries (Modena, Reggio Emilia, Parma, Ferrara)

  • Histologicallyconfirmed diagnosis of HL between 1 January 2006 and 31 December 2008, age between 18 and 75 years, and human immunodeficiency virus (HIV) negativity

Exclusion criteria

  • Missing data

Treatment

  • N = 116 (85%) participants received ABVD chemotherapy, 11 participants (8%) received intensified regimens such as BEACOPP or COPP/EBV/CAD, six participants (4%) received chemotherapy without anthracycline such as VBM or MOPP, and three participants (3%) received other therapies such as radiotherapy alone

Primary outcome measure(s)

  • Failure‐free survival

  • Overall survival

Medvedovskaya 2016

Notes

Title: The impact of outcome of interim PET‐CT on advanced Hodgkin lymphoma treated with EACOPP‐14.

(Conference abstract)

Aim

  • To assess the role of interim PET‐CT and compare it with PET‐CT results after the end of treatment in individuals with advanced stage classic HL

Study design

  • Not reported

Country/treatment centre(s)

  • Russia

Number of included participants

  • 114

Inclusion criteria

  • Newly diagnosed classic HL

Exclusion criteria

  • None

Treatment

  • 6 cycles of EACOPP‐14 with or without RT

Primary outcome measure(s)

  • Complete metabolic response

Molnar 2011

Notes

Title: The value of interim 18F‐FDG PET‐CT in Hodgkin lymphoma.

(Conference abstract)

Aim

  • To summarise our experience with 18F‐FDG PET‐CT in HL

Study design

  • Retrospective, November 2006 to January 2010

Country/treatment centre(s)

  • Hungary (National Institute of Oncology, Budapest)

Number of included participants

  • 60

Inclusion criteria

  • Not reported

Exclusion criteria

  • Not reported

Treatment

  • ABVD or BEACOPPesc, with or without RT

Primary outcome measure(s)

  • Prognostic value

Molnar 2011a

Notes

Title: Interim FDG PET‐CT examinations in advanced stage Hodgkin lymphoma.

(Conference abstract)

Aim

  • To summarise our experience with 18F‐FDG PET‐CT in interim staging

Study design

  • Retrospective, November 2007 to January 2010

Country/treatment centre(s)

  • Hungary (National Institute of Oncology, Budapest)

Number of included participants

  • 19

Inclusion criteria

  • Not reported

Exclusion criteria

  • Not reported

Treatment

  • ABVD or BEACOPPesc, with or without RT

Primary outcome measure(s)

  • Prognostic value

Moreira 2013

Notes

Title: Prognostic value of interim vs. end‐of‐treatment PET scan in Hodgkin's lymphoma.

(Confernece abstract)

Aim

  • To evaluate the prognostic value of interim PET scan (PET2) and end‐of‐treatment PET (PET6) in the outcome of individuals with HL

Study design

  • Retrospective, January 2004 to December 2011

Country/treatment centre(s)

  • Portugal (Porto, single‐centre)

Number of included participants

  • 261

Inclusion criteria

  • Diagnosed HL

Exclusion criteria

  • PET‐guided treatment adaptation

Treatment

  • ABVD, BEACOPPesc or CVP/CEB

Primary outcome measure(s)

  • Complete remission

  • Overall survival

  • Progression‐free survival

Perrone 2009

Notes

Title: Role of positron emission tomography (PET) after 2 and 4 courses of chemotherapy in patients with Hodgkin's lymphoma: A single center experience.

(Conference abstract)

Aim

  • To investigate the value of PET performed after 2 (PET2) and 4 (PET4) cycles of therapy for the management of patients with HL

Study design

  • Not reported, September 2006 to September 2008

Country/treatment centre(s)

  • Italy (University of Bari)

Number of included participants

  • 26

Inclusion criteria

  • Newly diagnosed HL

Exclusion criteria

  • None

Treatment

  • ABVD

Primary outcome measure(s)

  • Complete remission

  • Partial remission

  • Progression‐free survival

Pophali 2014

Notes

Title: Bulky disease does not adversely affect overall survival in early stage Hodgkin lymphoma: Role of interim PET and possible omission of radiotherapy in select patients.

(Conference abstract)

Aim

  • To assess the impact of disease bulk, interim PET and treatment modality on outcomes

Study design

  • Retrospective, 1995 to 2011

Country/treatment centre(s)

  • USA (Cleveland Clinic, Cleveland OH)

Number of included participants

  • 121

Inclusion criteria

  • Previously untreated HL, early stages (I and II)

Exclusion criteria

  • Missing clinical data

Treatment

  • ABVD or other chemotherapy (not specified)

Primary outcome measure(s)

  • Overall survival

  • Progression‐free survival

Rusconi 2010

Notes

Title: Baseline and dynamic prognostic factors in newly diagnosed classical Hodgkin's lymphoma.

(Conference abstract)

Aim

  • To identify characteristics, both at baseline and during therapy, predictive for survival outcomes in HL

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy (Niguarda Hospital, Milan)

Number of included participants

  • 105

Inclusion criteria

  • Diagnosed HL

Exclusion criteria

  • None

Treatment

  • 3 to 8 cycles of ABVD with or without IFRT

Primary outcome measure(s)

  • Overall survival

  • Event‐free survival

  • Relapse‐free survival

Spallino 2017

Notes

Title: The Deauville criteria and QPET as prognostic factors in interim PET in adult Hodgkin lymphoma: A single centre experience.

(Conference abstract)

Aim

  • To explore the prognostic role of iPET in individuals with HL by correlating Deauville criteria and qPET to DFS and OS

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy

Number of included participants

  • 131

Inclusion criteria

  • Newly diagnosed HL, disease stages I to IV

Exclusion criteria

  • None

Treatment

  • Not reported

Primary outcome measure(s)

  • Overall survival

  • Disease‐free survival

Yaghmour 2012

Notes

Title: PET‐negative at 2, 3 or 4 cycles of ABVD in Hodgkin's lymphoma is still good.

(Conference abstract)

Aim

  • To assess the prognostic value of anytime negative PET scan in the course of first line treatment in individuals with HL receiving ABVD

Study design

  • Retrospective

Country/treatment centre(s)

  • USA (Henry Ford Health System, Detroit MI)

Number of included participants

  • 32

Inclusion criteria

  • Newly diagnosed HL

Exclusion criteria

  • Not reported

Treatment

  • ABVD

Primary outcome measure(s)

  • Overall survival

Zanoni 2011

Notes

Title: The predictive value of interim PET and immunohistochemical markers in Hodgkin lymphoma (HL).

(Conference abstract)

Aim

  • To compare iPET with a series of histological and immunohistochemical parameters obtained on tissue‐micro‐arrays as possible predictive factors

Study design

  • Retrospective

Country/treatment centre(s)

  • Italy (Bologna)

Number of included participants

  • 209

Inclusion criteria

  • Biopsy‐proven HL, complete clinical and iPET data

Exclusion criteria

  • None

Treatment

  • Not reported

Primary outcome measure(s)

  • Overall survival

  • Progression‐free survival

ABVD: adriamycin/doxorubicin, bleomycin, vinblastine and dacarbazine; BEACOPP: bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine and prednisone; EBVD: epirubicin, bleomycin, vinblastine and dacarbazine; FDG: [18F]‐fluorodeoxy‐D‐glucose; HL: Hodgkin lymphoma; IF‐RT: involved‐field radiation therapy; iPET: interim positron emission tomography; MOPP: mustargen, Oncovin, procarbazine and prednisone; NPV: negative predictive value; PET: positron emission tomography; PET‐CT: positron emission tomography computed tomography; RT: radiotherapy.

Characteristics of ongoing studies [ordered by study ID]

NCT00736320

Study name

HD16 for Early Stages ‐ Treatment optimisation trial in the first‐line treatment of early stage Hodgkin lymphoma; treatment stratification by means of FDG‐PET

Starting date

November 2009

Contact information

Prof. Dr. Andreas Engert, University of Cologne, Germany

Notes

Study design

  • Randomised clinical trial (phase III) including 1150 participants with HL

Country/treatment centre

  • 1st Department of Medicine, Cologne University Hospital, Cologne, Germany

Number of included participants

  • Total: 1150

Inclusion criteria

  • Hodgkin lymphoma

  • Adults (18 to 75 years)

  • CS I and II without risk factors (large mediastinal mass (> 1/3 of maximum transverse thorax diameter), extranodal involvement, elevated ESR, three or more involved nodal areas)

  • Written informed consent

Exclusion criteria

  • Leukocytes < 3000/µl

  • Platelets < 100000/µl

  • Hodgkin lymphoma as composite lymphoma

  • Activity index (WHO) > 2

Arms and interventions

  • Active comparator (A): two cycles ABVD followed by 20 Gy IF‐RT, irrespective of FDG‐PET results after chemotherapy

  • Expertimental (B): two cycles ABVD followed by 20 Gy IF‐RT if FDG‐PET is positive after chemotherapy; 2 cycles ABVD and treatment stop if FDG‐PET is negative after chemotherapy

Primary outcome measure(s)

  • Progression‐free survival

  • Time frame: five years

Secondary outcome measure(s)

  • Overall survival

  • Acute toxicity

  • Late toxicity

  • Complete response rate

  • Time frame: five years

Estimated study completion date

  • May 2020

ESR: erythrocyte sedimentation rate; FDG: [18F]‐fluorodeoxy‐D‐glucose; HL: Hodgkin lymphoma; IF‐RT :involved‐field radiation therapy; MDCT: multi detector computed tomography; WHO: World Health Organization

Data and analyses

Open in table viewer
Comparison 1. Univariable comparison of PET+ve vs. PET‐ve

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Overall survival Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

Analysis 1.1

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 1: Overall survival

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 1: Overall survival

1.2 Progression‐free survival Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

Analysis 1.2

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 2: Progression‐free survival

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 2: Progression‐free survival

Open in table viewer
Comparison 2. Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 OS by radiotherapy Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

Analysis 2.1

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 1: OS by radiotherapy

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 1: OS by radiotherapy

2.1.1 Involved node and/or site

3

548

Hazard Ratio (IV, Random, 95% CI)

3.45 [1.22, 9.72]

2.1.2 involved field

4

428

Hazard Ratio (IV, Random, 95% CI)

12.75 [4.98, 32.68]

2.1.3 not specified

2

826

Hazard Ratio (IV, Random, 95% CI)

2.80 [1.17, 6.67]

2.2 OS by study design Show forest plot

8

1717

Hazard Ratio (IV, Random, 95% CI)

4.63 [2.43, 8.80]

Analysis 2.2

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 2: OS by study design

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 2: OS by study design

2.2.1 Prospective

3

406

Hazard Ratio (IV, Random, 95% CI)

5.35 [1.07, 26.68]

2.2.2 Retrospective

4

589

Hazard Ratio (IV, Random, 95% CI)

7.12 [3.14, 16.14]

2.2.3 RCT

1

722

Hazard Ratio (IV, Random, 95% CI)

2.60 [1.03, 6.56]

2.3 OS by chemotherapy Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

Analysis 2.3

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 3: OS by chemotherapy

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 3: OS by chemotherapy

2.3.1 ABVD

5

801

Hazard Ratio (IV, Random, 95% CI)

5.19 [2.11, 12.72]

2.3.2 ABVD and/or other

3

279

Hazard Ratio (IV, Random, 95% CI)

10.30 [1.71, 62.13]

2.3.3 BEACOPP

1

722

Hazard Ratio (IV, Random, 95% CI)

2.60 [1.03, 6.56]

2.4 OS for PET/CT vs PET Show forest plot

8

1706

Hazard Ratio (IV, Random, 95% CI)

5.01 [2.50, 10.02]

Analysis 2.4

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 4: OS for PET/CT vs PET

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 4: OS for PET/CT vs PET

2.4.1 PET/CT

5

595

Hazard Ratio (IV, Random, 95% CI)

4.70 [1.86, 11.86]

2.4.2 PET only

3

1111

Hazard Ratio (IV, Random, 95% CI)

6.99 [1.58, 30.90]

2.5 OS by disease stage Show forest plot

9

1802

Odds Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

Analysis 2.5

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 5: OS by disease stage

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 5: OS by disease stage

2.5.1 Stages I and II with A and B symptoms

1

96

Odds Ratio (IV, Random, 95% CI)

9.21 [0.71, 120.03]

2.5.2 All stages

7

984

Odds Ratio (IV, Random, 95% CI)

6.28 [2.62, 15.05]

2.5.3 Advanced

1

722

Odds Ratio (IV, Random, 95% CI)

2.60 [1.03, 6.56]

2.6 Timing of interim PET Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

Analysis 2.6

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

2.6.1 PET2

6

1495

Hazard Ratio (IV, Random, 95% CI)

3.53 [1.97, 6.32]

2.6.2 Other (including mixed)

3

307

Hazard Ratio (IV, Random, 95% CI)

20.13 [5.04, 80.38]

2.7 OS by HR type of estimation Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

Analysis 2.7

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 7: OS by HR type of estimation

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 7: OS by HR type of estimation

2.7.1 precise

7

1638

Hazard Ratio (IV, Random, 95% CI)

5.70 [2.60, 12.48]

2.7.2 Imprecise

2

164

Hazard Ratio (IV, Random, 95% CI)

3.60 [0.89, 14.64]

Open in table viewer
Comparison 3. Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 PFS by study design Show forest plot

13

1349

Hazard Ratio (IV, Random, 95% CI)

5.66 [4.02, 7.97]

Analysis 3.1

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 1: PFS by study design

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 1: PFS by study design

3.1.1 prospective

3

357

Hazard Ratio (IV, Random, 95% CI)

3.95 [2.23, 7.00]

3.1.2 retrospective

8

827

Hazard Ratio (IV, Random, 95% CI)

6.85 [4.66, 10.08]

3.1.3 RCT

2

165

Hazard Ratio (IV, Random, 95% CI)

6.21 [2.87, 13.42]

3.2 PFS by chemotherapy Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

Analysis 3.2

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 2: PFS by chemotherapy

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 2: PFS by chemotherapy

3.2.1 ABVD

7

945

Hazard Ratio (IV, Random, 95% CI)

5.13 [3.18, 8.27]

3.2.2 ABVD and/or other

4

265

Hazard Ratio (IV, Random, 95% CI)

7.07 [3.40, 14.70]

3.2.3 other NON‐ABVD chemo

3

869

Hazard Ratio (IV, Random, 95% CI)

3.64 [1.83, 7.24]

3.3 PFS for PET/CT vs PET Show forest plot

13

1983

Hazard Ratio (IV, Random, 95% CI)

5.08 [3.57, 7.21]

Analysis 3.3

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 3: PFS for PET/CT vs PET

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 3: PFS for PET/CT vs PET

3.3.1 PET/CT

8

707

Hazard Ratio (IV, Random, 95% CI)

6.03 [3.68, 9.90]

3.3.2 PET only

5

1276

Hazard Ratio (IV, Random, 95% CI)

4.06 [2.33, 7.08]

3.4 PFS by disease stage Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

Analysis 3.4

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 4: PFS by disease stage

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 4: PFS by disease stage

3.4.1 Stages I and II with A and B symptoms

2

184

Hazard Ratio (IV, Random, 95% CI)

3.88 [1.54, 9.83]

3.4.2 All stages

11

1173

Hazard Ratio (IV, Random, 95% CI)

5.81 [3.93, 8.57]

3.4.3 Advanced

1

722

Hazard Ratio (IV, Random, 95% CI)

2.27 [1.35, 3.82]

3.5 PFS by radiotherapy Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

Analysis 3.5

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 5: PFS by radiotherapy

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 5: PFS by radiotherapy

3.5.1 Involved node and/or site

5

651

Hazard Ratio (IV, Random, 95% CI)

5.35 [2.94, 9.75]

3.5.2 Involved field

6

514

Hazard Ratio (IV, Random, 95% CI)

7.06 [4.15, 12.00]

3.5.3 Not specified

2

826

Hazard Ratio (IV, Random, 95% CI)

2.97 [1.48, 5.98]

3.5.4 None

1

88

Hazard Ratio (IV, Random, 95% CI)

5.09 [1.95, 13.29]

3.6 Timing of interim PET Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

Analysis 3.6

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

3.6.1 PET2

9

1677

Hazard Ratio (IV, Random, 95% CI)

4.68 [3.14, 6.98]

3.6.2 Other (including mixed)

5

402

Hazard Ratio (IV, Random, 95% CI)

6.32 [3.40, 11.75]

3.7 PFS by HR type of estimation Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

Analysis 3.7

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 7: PFS by HR type of estimation

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 7: PFS by HR type of estimation

3.7.1 precise

9

1450

Hazard Ratio (IV, Random, 95% CI)

4.69 [2.84, 7.73]

3.7.2 Imprecise

5

629

Hazard Ratio (IV, Random, 95% CI)

5.66 [3.65, 8.77]

Study flow diagram according to PRISMA

Figuras y tablas -
Figure 1

Study flow diagram according to PRISMA

'Risk of bias' assessment according to QUIPS (Quality in Prognostic Studies) by outcome.

Figuras y tablas -
Figure 2

'Risk of bias' assessment according to QUIPS (Quality in Prognostic Studies) by outcome.

Forest plot of comparison: 1 Univariable comparison of PET+ve vs. PET‐ve, outcome: 1.1 Overall survival

Figuras y tablas -
Figure 3

Forest plot of comparison: 1 Univariable comparison of PET+ve vs. PET‐ve, outcome: 1.1 Overall survival

Forest plot of comparison: 1 Univariable comparison of PET+ve vs. PET‐ve, outcome: 1.2 Progression‐free survival

Figuras y tablas -
Figure 4

Forest plot of comparison: 1 Univariable comparison of PET+ve vs. PET‐ve, outcome: 1.2 Progression‐free survival

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 1: Overall survival

Figuras y tablas -
Analysis 1.1

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 1: Overall survival

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 2: Progression‐free survival

Figuras y tablas -
Analysis 1.2

Comparison 1: Univariable comparison of PET+ve vs. PET‐ve, Outcome 2: Progression‐free survival

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 1: OS by radiotherapy

Figuras y tablas -
Analysis 2.1

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 1: OS by radiotherapy

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 2: OS by study design

Figuras y tablas -
Analysis 2.2

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 2: OS by study design

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 3: OS by chemotherapy

Figuras y tablas -
Analysis 2.3

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 3: OS by chemotherapy

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 4: OS for PET/CT vs PET

Figuras y tablas -
Analysis 2.4

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 4: OS for PET/CT vs PET

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 5: OS by disease stage

Figuras y tablas -
Analysis 2.5

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 5: OS by disease stage

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

Figuras y tablas -
Analysis 2.6

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 7: OS by HR type of estimation

Figuras y tablas -
Analysis 2.7

Comparison 2: Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve, Outcome 7: OS by HR type of estimation

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 1: PFS by study design

Figuras y tablas -
Analysis 3.1

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 1: PFS by study design

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 2: PFS by chemotherapy

Figuras y tablas -
Analysis 3.2

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 2: PFS by chemotherapy

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 3: PFS for PET/CT vs PET

Figuras y tablas -
Analysis 3.3

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 3: PFS for PET/CT vs PET

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 4: PFS by disease stage

Figuras y tablas -
Analysis 3.4

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 4: PFS by disease stage

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 5: PFS by radiotherapy

Figuras y tablas -
Analysis 3.5

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 5: PFS by radiotherapy

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

Figuras y tablas -
Analysis 3.6

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 6: Timing of interim PET

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 7: PFS by HR type of estimation

Figuras y tablas -
Analysis 3.7

Comparison 3: Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve, Outcome 7: PFS by HR type of estimation

Summary of findings 1. Comparison of interim PET‐negative and interim PET‐positive individuals with Hodgkin Lymphoma

Comparison of interim PET‐positive and interim PET‐negative participants with Hodgkin lymphoma

Population: Individuals with Hodgkin lymphoma
Setting: Eleven studies recruited participants from a total of 28 haemato‐oncology treatment centres/hospitals in Brazil (N = 1), China (N = 1), Denmark (N = 4), France (N = 4), Italy (N = 3), Poland (N = 11), UK (N = 2) and the USA (N = 2). One study (Straus 2011) included participants from 29 institutions, but did not report the countries. One study (Simon 2016) reported the country (Hungary) but not the number of centres. One multi‐centre study (Hutchings 2014) recruited participants from four countries (USA, Italy, Poland and Denmark). One RCT (Kobe 2018) included participants from 301 hospitals and private practices in Germany, Switzerland, Austria, the Netherlands, and the Czech Republic.

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

№ of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with Interim PET‐negative

Risk with Interim PET‐positive

Overall survival

Follow up: 3 years

Low

HR 5.09
(2.64 to 9.81)

1802
(9 studies)

⊕⊕⊕⊝
MODERATE 2 3 4

900 per 1.000 1

585 per 1.0001
(356 to 757)

High

980 per 1.000 1

902 per 1.0001
(820 to 948)

Progression‐free survival

Follow up: 3 years

Low

HR 4.90
(3.47 to 6.90)

2079
(14 studies)

⊕⊝⊝⊝
VERY LOW6 7 8

850 per 1.000 5

451 per 1.000 5
(326 to 569)

High

940 per 1.000 5

738 per 1.000 5
(653 to 807)

Adverse events associated with PET ‐ not reported

No study measured PET‐associated adverse events.

Overall survival (adjusted effect estimate)

Two studies reported an adjusted effect estimate for overall survival after interim PET2: a hazard ratio of 3.2 (95% CI 1.3 to 8.4, P = 0.02) (Kobe 2018) and 11.51 (95% CI 3.14 to 42.86, P < 0.001) (Simon 2015) indicates the independent prognostic value of interim PET over and above other clinically relevant prognostic factors.

843
(2 studies)

⊕⊕⊕⊝
MODERATE 9

Progression‐free survival (adjusted effect estimate)

Eight studies conducted a multivariable analysis to test the independent prognostic value of interim PET over and above other clinically relevant prognostic factors. Four of these studies reported a hazard ratio as the adjusted effect estimate, of which the value ranges from 2.4 to 36.89, indicating the independent prognostic value of interim PET2.10

996
(4 studies)10

⊕⊕⊝⊝
LOW 11 12

*The survival in the PET‐positive group (and its 95% confidence interval) is based on the assumed survival in the PET‐negative group.

CI: Confidence interval; HR: Hazard ratio; PET: positron emission tomography

GRADE Working Group grades of evidence
High certainty: We are very confident that the true effect lies close to that of the estimate of the effect
Moderate certainty: 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 certainty: Our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect
Very low certainty: We have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect

1 The assumed event‐free survival in the control group is based on the survival rate of the interim PET‐negative participants at 3 years in the studies included (the lowest survival rate from Cerci 2010 and the highest survival rate from Kobe 2018).

2 High risk of bias in seven studies for the domain 'other prognostic factors (covariates)', and high risk of bias in three studies for the domain 'statistical analysis and reporting'. Downgraded by 1 point for risk of bias.

3 For one study we used the reported hazard ratio. For seven studies we had to estimate the hazard ratio and for one study we re‐calculated it (Trivella 2006). Downgraded by 1 point for imprecision.

4 Upgraded by one point due to the large effect showing the large difference between interim PET‐negative and interim PET‐positive participants (HR 5.09, CI 2.64 to 9.81).

5 The assumed event‐free survival in the control group is based on the survival rate of the interim PET‐negative participants at 3 years in the studies included (the lowest survival rate from Rossi 2014 and the highest survival rate from Kobe 2018).

6 High risk of bias in eight studies for the domain 'other prognostic factors (covariates)', and high risk of bias in six studies for the domain 'statistical analysis and reporting'. Downgraded by 1 point for risk of bias.

7The definition of PFS varied across studies, downgraded by 1 point for inconsistency

8 For three studies we used the reported hazard ratio. For ten studies we had to estimate the value, and for one study we had to re‐calculate it (Trivella 2006). Downgraded by 1 point for imprecision.

9 High risk of bias for the domains 'other prognostic factors (covariates)' and statistical analysis and reporting for one study (Simon 2016). Downgraded by 1 point for risk of bias.

10Hutchings 2006; Kobe 2018; Mesguich 2016; Simon 2016.

11 High risk of bias for the domains 'other prognostic factors (covariates)' and statistical analysis and reporting for one study (Simon 2016). Also high risk of bias for the domain study participation in one study (Hutchings 2006). Downgraded by 1 point for risk of bias.

12 Studies included a heterogenous set of covariates in the adjusted analyses. Downgraded by 1 point for inconsistency.

Figuras y tablas -
Summary of findings 1. Comparison of interim PET‐negative and interim PET‐positive individuals with Hodgkin Lymphoma
Comparison 1. Univariable comparison of PET+ve vs. PET‐ve

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1.1 Overall survival Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

1.2 Progression‐free survival Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

Figuras y tablas -
Comparison 1. Univariable comparison of PET+ve vs. PET‐ve
Comparison 2. Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

2.1 OS by radiotherapy Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

2.1.1 Involved node and/or site

3

548

Hazard Ratio (IV, Random, 95% CI)

3.45 [1.22, 9.72]

2.1.2 involved field

4

428

Hazard Ratio (IV, Random, 95% CI)

12.75 [4.98, 32.68]

2.1.3 not specified

2

826

Hazard Ratio (IV, Random, 95% CI)

2.80 [1.17, 6.67]

2.2 OS by study design Show forest plot

8

1717

Hazard Ratio (IV, Random, 95% CI)

4.63 [2.43, 8.80]

2.2.1 Prospective

3

406

Hazard Ratio (IV, Random, 95% CI)

5.35 [1.07, 26.68]

2.2.2 Retrospective

4

589

Hazard Ratio (IV, Random, 95% CI)

7.12 [3.14, 16.14]

2.2.3 RCT

1

722

Hazard Ratio (IV, Random, 95% CI)

2.60 [1.03, 6.56]

2.3 OS by chemotherapy Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

2.3.1 ABVD

5

801

Hazard Ratio (IV, Random, 95% CI)

5.19 [2.11, 12.72]

2.3.2 ABVD and/or other

3

279

Hazard Ratio (IV, Random, 95% CI)

10.30 [1.71, 62.13]

2.3.3 BEACOPP

1

722

Hazard Ratio (IV, Random, 95% CI)

2.60 [1.03, 6.56]

2.4 OS for PET/CT vs PET Show forest plot

8

1706

Hazard Ratio (IV, Random, 95% CI)

5.01 [2.50, 10.02]

2.4.1 PET/CT

5

595

Hazard Ratio (IV, Random, 95% CI)

4.70 [1.86, 11.86]

2.4.2 PET only

3

1111

Hazard Ratio (IV, Random, 95% CI)

6.99 [1.58, 30.90]

2.5 OS by disease stage Show forest plot

9

1802

Odds Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

2.5.1 Stages I and II with A and B symptoms

1

96

Odds Ratio (IV, Random, 95% CI)

9.21 [0.71, 120.03]

2.5.2 All stages

7

984

Odds Ratio (IV, Random, 95% CI)

6.28 [2.62, 15.05]

2.5.3 Advanced

1

722

Odds Ratio (IV, Random, 95% CI)

2.60 [1.03, 6.56]

2.6 Timing of interim PET Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

2.6.1 PET2

6

1495

Hazard Ratio (IV, Random, 95% CI)

3.53 [1.97, 6.32]

2.6.2 Other (including mixed)

3

307

Hazard Ratio (IV, Random, 95% CI)

20.13 [5.04, 80.38]

2.7 OS by HR type of estimation Show forest plot

9

1802

Hazard Ratio (IV, Random, 95% CI)

5.09 [2.64, 9.81]

2.7.1 precise

7

1638

Hazard Ratio (IV, Random, 95% CI)

5.70 [2.60, 12.48]

2.7.2 Imprecise

2

164

Hazard Ratio (IV, Random, 95% CI)

3.60 [0.89, 14.64]

Figuras y tablas -
Comparison 2. Subgroups in univariable comparison of OS: PET+ve vs. PET‐ve
Comparison 3. Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

3.1 PFS by study design Show forest plot

13

1349

Hazard Ratio (IV, Random, 95% CI)

5.66 [4.02, 7.97]

3.1.1 prospective

3

357

Hazard Ratio (IV, Random, 95% CI)

3.95 [2.23, 7.00]

3.1.2 retrospective

8

827

Hazard Ratio (IV, Random, 95% CI)

6.85 [4.66, 10.08]

3.1.3 RCT

2

165

Hazard Ratio (IV, Random, 95% CI)

6.21 [2.87, 13.42]

3.2 PFS by chemotherapy Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

3.2.1 ABVD

7

945

Hazard Ratio (IV, Random, 95% CI)

5.13 [3.18, 8.27]

3.2.2 ABVD and/or other

4

265

Hazard Ratio (IV, Random, 95% CI)

7.07 [3.40, 14.70]

3.2.3 other NON‐ABVD chemo

3

869

Hazard Ratio (IV, Random, 95% CI)

3.64 [1.83, 7.24]

3.3 PFS for PET/CT vs PET Show forest plot

13

1983

Hazard Ratio (IV, Random, 95% CI)

5.08 [3.57, 7.21]

3.3.1 PET/CT

8

707

Hazard Ratio (IV, Random, 95% CI)

6.03 [3.68, 9.90]

3.3.2 PET only

5

1276

Hazard Ratio (IV, Random, 95% CI)

4.06 [2.33, 7.08]

3.4 PFS by disease stage Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

3.4.1 Stages I and II with A and B symptoms

2

184

Hazard Ratio (IV, Random, 95% CI)

3.88 [1.54, 9.83]

3.4.2 All stages

11

1173

Hazard Ratio (IV, Random, 95% CI)

5.81 [3.93, 8.57]

3.4.3 Advanced

1

722

Hazard Ratio (IV, Random, 95% CI)

2.27 [1.35, 3.82]

3.5 PFS by radiotherapy Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

3.5.1 Involved node and/or site

5

651

Hazard Ratio (IV, Random, 95% CI)

5.35 [2.94, 9.75]

3.5.2 Involved field

6

514

Hazard Ratio (IV, Random, 95% CI)

7.06 [4.15, 12.00]

3.5.3 Not specified

2

826

Hazard Ratio (IV, Random, 95% CI)

2.97 [1.48, 5.98]

3.5.4 None

1

88

Hazard Ratio (IV, Random, 95% CI)

5.09 [1.95, 13.29]

3.6 Timing of interim PET Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

3.6.1 PET2

9

1677

Hazard Ratio (IV, Random, 95% CI)

4.68 [3.14, 6.98]

3.6.2 Other (including mixed)

5

402

Hazard Ratio (IV, Random, 95% CI)

6.32 [3.40, 11.75]

3.7 PFS by HR type of estimation Show forest plot

14

2079

Hazard Ratio (IV, Random, 95% CI)

4.90 [3.47, 6.90]

3.7.1 precise

9

1450

Hazard Ratio (IV, Random, 95% CI)

4.69 [2.84, 7.73]

3.7.2 Imprecise

5

629

Hazard Ratio (IV, Random, 95% CI)

5.66 [3.65, 8.77]

Figuras y tablas -
Comparison 3. Subgroups in univariable comparison of PFS: PET+ve vs. PET‐ve