Scolaris Content Display Scolaris Content Display

Frühe palliative Versorgung für Erwachsene mit fortgeschrittener Krebserkrankung

Contraer todo Desplegar todo

Referencias

Bakitas 2009 {published and unpublished data}

Bakitas M, Lyons KD, Hegel MT, Ahles T. Oncologists' perspectives on concurrent palliative care in a National Cancer Institute‐designated comprehensive cancer center. Palliative & Supportive Care 2013;11:415‐23. [DOI: 10.1017/S1478951512000673]CENTRAL
Bakitas M, Lyons KD, Hegel MT, Balan S, Barnett KN, Brokaw FC, et al. The project ENABLE II randomized controlled trial to improve palliative care for rural patients with advanced cancer: baseline findings, methodological challenges, and solutions. Palliative & Supportive Care 2009;7:75‐86. [DOI: 10.1017/S1478951509000108]CENTRAL
Bakitas M, Lyons KD, Hegel MT, Balan S, Brokaw FC, Seville J, et al. Effects of a palliative care intervention on clinical outcomes in patients with advanced cancer: the Project ENABLE II randomized controlled trial. JAMA 2009;302(7):741‐9. [DOI: 10.1001/jama.2009.1198]CENTRAL
Bakitas M, Stevens M, Ahles T, Kirn M, Skalla K, Kane N, et al. Project ENABLE: a palliative care demonstration project for advanced cancer patients in three settings. Journal of Palliative Medicine 2004;7:363‐72. [DOI: 10.1089/109662104773709530]CENTRAL
Maloney C, Lyons KD, Li Z, Hegel M, Ahles TA, Bakitas M. Patient perspectives on participation in the ENABLE II randomized controlled trial of a concurrent oncology palliative care intervention: benefits and burdens. Palliative Medicine 2013;27:375‐83. [DOI: 10.1177/0269216312445188]CENTRAL
O'Hara RE, Hull JG, Lyons KD, Bakitas M, Hegel MT, Li Z, et al. Impact on caregiver burden of a patient‐focused palliative care intervention for patients with advanced cancer. Palliative & Supportive Care 2010;8:395‐404. [DOI: 10.1017/S1478951510000258]CENTRAL

Bakitas 2015 {published and unpublished data}

Bakitas MA, Tosteson TD, Li Z, Lyons KD, Hull JG, Li Z, et al. Early versus delayed initiation of concurrent palliative oncology care: patient outcomes in the ENABLE III randomized controlled trial. Journal of Clinical Oncology 2015;33(13):1438‐45. [DOI: 10.1200/JCO.2014.58.6362]CENTRAL
Dionne‐Odom J, Hull J, Martin M, Akyar I, Lyons K, Tosteson T, et al. The association between family caregiver burden and the survival of advanced cancer patients. Psycho‐Oncology 2015;24:57. [DOI: 10.1002/pon.3873]CENTRAL
Dionne‐Odom J, Raju D, Hull J, Imatullah A, Bakitas M. Characteristics and outcomes of persons with advanced cancer associated with having a family caregiver: a classification tree analysis. Journal of Pain and Symptom Management 2015;49:419‐20. [DOI: 10.1016/j.jpainsymman.2014.11.205]CENTRAL
Dionne‐Odom JN, Azuero A, Lyons KD, Hull JG, Tosteson T, Li Z. Benefits of early versus delayed palliative care to informal family caregivers of patients with advanced cancer: outcomes from the ENABLE III randomized controlled trial. Journal of Clinical Oncology 2015;33:1446‐52. [DOI: 10.1200/JCO.2014.58.7824]CENTRAL
Dionne‐Odom JN, Hull JG, Martin MY, Lyons KD, Prescott AT, Tosteson T, et al. Associations between advanced cancer patients' survival and family caregiver presence and burden. Cancer Medicine 2016;5:853‐62. [DOI: 10.1002/cam4.653]CENTRAL

Maltoni 2016 {published and unpublished data}

Maltoni M, Scarpi E, Dall'agata M, Schiavon S, Biasini C, Codeca C, et al. Systematic versus on‐demand early palliative care: a randomised clinical trial assessing quality of care and treatment aggressiveness near the end of life. European Journal of Cancer 2016;69:110‐8. [DOI: 10.1016/j.ejca.2016.10.004]CENTRAL
Maltoni M, Scarpi E, Dall’Agata M, Zagonel V, Berte R, Ferrari D, et al. Systematic versus on‐demand early palliative care: results from a multicentre, randomised clinical trial. European Journal of Cancer 2016;65:61‐8. [DOI: 10.1016/j.ejca.2016.06.007]CENTRAL

McCorkle 2015 {published and unpublished data}

McCorkle R, Jeon S, Ercolano E, Lazenby M, Reid A, Davies M, et al. An advanced practice nurse coordinated multidisciplinary intervention for patients with late‐stage cancer: a cluster randomized trial. Journal of Palliative Medicine 2015;18:962‐9. [DOI: 10.1089/jpm.2015.0113]CENTRAL

Tattersall 2014 {published and unpublished data}

Tattersall MHN, Martin A, Devine R, Ryan J, Jansen J, Hastings L, et al. Early contact with palliative care services: a randomized trial in patients with newly detected incurable metastatic cancer. Palliative Care & Medicine 2014;4(1):170. [DOI: 10.4172/2165‐7386.1000170]CENTRAL

Temel 2010 {published and unpublished data}

Back AL, Park ER, Greer JA, Jackson VA, Jacobsen JC, Gallagher ER, et al. Clinician roles in early integrated palliative care for patients with advanced cancer: a qualitative study. Journal of Palliative Medicine 2014;17:1244‐8. [DOI: 10.1089/jpm.2014.0146]CENTRAL
Fujisawa D, Temel JS, Traeger L, Greer JA, Lennes IT, Mimura M, et al. Psychological factors at early stage of treatment as predictors of receiving chemotherapy at the end of life. Psycho‐Oncology 2015;24:1731‐7. [DOI: 10.1002/pon.3840]CENTRAL
Greer JA, Pirl WF, Jackson VA, Muzikansky A, Lennes IT, Heist RS, et al. Effect of early palliative care on chemotherapy use and end‐of‐life care in patients with metastatic non‐small‐cell lung cancer. Journal of Clinical Oncology 2012;30:394‐400. [DOI: 10.1200/JCO.2011.35.7996]CENTRAL
Jacobsen J, Jackson V, Dahlin C, Greer J, Perez‐Cruz P, Billings JA, et al. Components of early outpatient palliative care consultation in patients with metastatic nonsmall cell lung cancer. Journal of Palliative Medicine 2011;14:459‐64. [DOI: 10.1089/jpm.2010.0382]CENTRAL
Nipp RD, Greer JA, El‐Jawahri A, Traeger L, Gallagher ER, Park ER, et al. Age and gender moderate the impact of early palliative care in metastatic non‐small cell lung cancer. The Oncologist 2016;21:119‐26. [DOI: 10.1634/theoncologist.2015‐0232]CENTRAL
Pirl WF, Greer JA, Traeger L, Jackson V, Lennes IT, Gallagher ER, et al. Depression and survival in metastatic non‐small‐cell lung cancer: effects of early palliative care. Journal of Clinical Oncology 2012;30:1310‐5. [DOI: 10.1200/JCO.2011.38.3166]CENTRAL
Pirl WF, Traeger L, Greer JA, Jackson V, Lennes IT, Gallagher E, et al. Depression, survival, and epidermal growth factor receptor genotypes in patients with metastatic non‐small cell lung cancer. Palliative & Supportive Care 2013;11:223‐9. [DOI: 10.1017/S1478951512001071]CENTRAL
Salman S, Idrees J, Idrees M, Anees M, Idrees F, Khattak AA. Early palliative care for patients with metastatic non‐small‐cell lung cancer in Khyber Pakhtunkhwa. Supportive Care in Cancer 2013;21:S178. [DOI: 10.1007/s00520‐013‐1798‐3]CENTRAL
Temel JS, Greer JA, Admane S, Gallagher ER, Jackson VA, Lynch TJ, et al. Longitudinal perceptions of prognosis and goals of therapy in patients with metastatic non‐small‐cell lung cancer: results of a randomized study of early palliative care. Journal of Clinical Oncology 2011;17:2319‐26. [DOI: 10.1200/JCO.2010.32.4459]CENTRAL
Temel JS, Greer JA, Muzikansky A, Gallagher ER, Admane S, Jackson VA, et al. Early palliative care for patients with metastatic non‐small‐cell lung cancer. New England Journal of Medicine 2010;363(8):733‐42. [DOI: 10.1056/NEJMoa1000678]CENTRAL
Temel JS, Jackson VA, Billings JA, Dahlin C, Block SD, Buss MK, et al. Phase II study: integrated palliative care in newly diagnosed advanced non‐small‐cell lung cancer patients. Journal of Clinical Oncology 2007;25:2377‐82. [DOI: 10.1200/JCO.2006.09.2627]CENTRAL
Yoong J, Park ER, Greer JA, Jackson VA, Gallagher ER, Pirl WF, et al. Early palliative care in advanced lung cancer: a qualitative study. JAMA Internal Medicine 2013;173:283‐90. [DOI: 10.1001/jamainternmed.2013.1874]CENTRAL

Zimmermann 2014 {published and unpublished data}

Follwell M, Burman D, Le LW, Wakimoto K, Seccareccia D, Bryson J, et al. Phase II study of an outpatient palliative care intervention in patients with metastatic cancer. Journal of Clinical Oncology 2009;27:206‐13. [DOI: 10.1200/JCO.2008.17.7568]CENTRAL
McDonald J, Swami N, Hannon B, Lo C, Pope A, Oza A, et al. Impact of early palliative care on caregivers of patients with advanced cancer: cluster randomised trial. Annals of Oncology 2016;[Epub ahead of print]:Sep 29. [DOI: 10.1093/annonc/mdw438]CENTRAL
Zimmermann C, Swami N, Krzyzanowska M, Hannon B, Leighl N, Oza A, et al. Early palliative care for patients with advanced cancer: a cluster‐randomised controlled trial. Lancet 2014;383(9930):1721‐30. [DOI: 10.1016/S0140‐6736(13)62416‐2]CENTRAL
Zimmermann C, Swami N, Krzyzanowska M, Leighl N, Rydall A, Rodin G, et al. Perceptions of palliative care among patients with advanced cancer and their caregivers. Canadian Medical Association Journal 2016;188:E217‐27. [DOI: 10.1503/cmaj.151171]CENTRAL

References to studies excluded from this review

Badr 2015 {published data only}

Badr H, Smith CB, Goldstein NE, Gomez JE, Redd WH. Dyadic psychosocial intervention for advanced lung cancer patients and their family caregivers: results of a randomized pilot trial. Cancer 2015;121:150‐8. [DOI: 10.1002/cncr.29009]CENTRAL

Brumley 2007 {published data only}

Brumley R, Enguidanos S, Jamison P, Seitz R, Morgenstern N, Saito S, et al. Increased satisfaction with care and lower costs: results of a randomized trial of in‐home palliative care. Journal of the American Geriatrics Society 2007;55:993‐1000. [DOI: 10.1111/j.1532‐5415.2007.01234.x]CENTRAL

Caruso 2014 {published data only}

Caruso R, Sabato S, Massarenti S, Nanni MG, Grassi L. The experience of cancer in advanced phases of illness: Italian CALM project. Psycho‐Oncology 2014;23(Suppl 3):S7‐0665. [DOI: 10.1111/j.1099‐1611.2014.3693]CENTRAL

Dyar 2012 {published data only}

Dyar S, Lesperance M, Shannon R, Sloan J, Colon‐Otero G. A nurse practitioner directed intervention improves the quality of life of patients with metastatic cancer: results of a randomized pilot study. Journal of Palliative Medicine 2012;8:890‐5. [DOI: 10.1089/jpm.2012.0014]CENTRAL

Ferrell 2015 {published data only}

Ferrell B, Sun V, Hurria A, Cristea M, Raz DJ, KIm JY, et al. Interdisciplinary palliative care for patients with lung cancer. Journal of Pain and Symptom Management 2015;50:758‐67. [DOI: 10.1016/j.jpainsymman.2015.07.005]CENTRAL

Gade 2008 {published data only}

Gade G, Venohr I, Conner D, McGrady K, Beane J, Richardson RH, et al. Impact of an inpatient palliative care team: a randomized control trial. Journal of Palliative Medicine 2008;11:180‐90. [DOI: 10.1089/jpm.2007.0055]CENTRAL

Grudzen 2016 {published data only}

Grudzen CR, Richardson LD, Johnson PN, Hu M, Wang B, Ortiz JM, et al. Emergency department‐initiated palliative care in advanced cancer: a randomized clinical trial. JAMA Oncology 2016;2:591‐8. [DOI: 10.1001/jamaoncol.2015.5252]CENTRAL
Kandarian B, Morrison RS, Richardson LD, Ortiz J, Grudzen CR. Emergency department‐initiated palliative care for advanced cancer patients: protocol for a pilot randomized controlled trial. Trials 2014;15:251. [DOI: 10.1186/1745‐6215‐15‐251]CENTRAL
Kistler EA, Sean Morrison R, Richardson LD, Ortiz JM, Grudzen CR. Emergency department‐triggered palliative care in advanced cancer: proof of concept. Academic Emergency Medicine 2015;22:237‐9. [DOI: 10.1111/acem.12573]CENTRAL

Jensen 2014 {published data only}

Jensen W, Baumann FT, Stein A, Bloch W, Bokemeyer C, de Wit M, et al. Exercise training in patients with advanced gastrointestinal cancer undergoing palliative chemotherapy: a pilot study. Support Care in Cancer 2014;22:1797‐80. [DOI: 10.1007/s00520‐014‐2139‐x]CENTRAL

Jordhoy 2001 {published data only}

Jordhøy MS, Fayers P, Loge JH, Ahlner‐Elmqvist M, Kaasa S. Quality of life in palliative cancer care: results from a cluster randomized trial. Journal of Clinical Oncology 2001;19:3884‐94. CENTRAL

Laing 1975 {published data only}

Laing AH, Berry RJ, Newman CR, Peto J. Treatment of inoperable carcinoma of bronchus. Lancet 1975;306(7946):1161‐4. [DOI: 10.1016/S0140‐6736(75)92654‐9]CENTRAL

Lloyd‐Williams 2013 {published data only}

Lloyd‐Williams M, Cobb M, O'Connor C, Dunn L, Shiels C. A pilot randomised controlled trial to reduce suffering and emotional distress in patients with advanced cancer. Journal of Affective Disorders 2013;148:141‐5. [DOI: 10.1016/j.jad.2012.11.013]CENTRAL

Mok 2012 {published data only}

Mok E, Lau KP, Lai T, Ching S. The meaning of life intervention for patients with advanced‐stage cancer: development and pilot study. Oncology Nursing Forum 2012;39:E480‐8. [DOI: 10.1188/12.ONF.E480‐E488]CENTRAL

NCT02311465 {published data only}

Responsible party: Gordon Bernard, MD (Professor of Medicine). A randomized study of early palliative care integrated with standard oncology care versus oncology care alone in patients with non‐colorectal gastrointestinal malignancies. Vanderbilt University, Nashville, Tennessee Study first received on December 4, 2014 on clinicaltrails.gov; Vol. withdrawn prior to enrolment. CENTRAL

Pantilat 2010 {published data only}

Pantilat SZ, O'Riordan DL, Dibble SL, Landefeld CS. Hospital‐based palliative medicine consultation: a randomized controlled trial. Archives of Internal Medicine 2010;170:2038‐40. [DOI: 10.1001/archinternmed.2010.460]CENTRAL

Rabow 2004 {published data only (unpublished sought but not used)}

Rabow MW, Dibble SL, Pantilat SZ, McPhee SJ. The comprehensive care team: a controlled trial of outpatient palliative medicine consultation. Archives of Internal Medicine 2004;164:83‐91. [PUBMED: 14718327]CENTRAL

Rummans 2006 {published data only}

Rummans TA, Clark MM, Sloan JA, Frost MH, Bostwick JM, Atherton PJ, et al. Impacting quality of life for patients with advanced cancer with a structured multidisciplinary intervention: a randomized controlled trial. Journal of Clinical Oncology 2006;24:635‐42. [DOI: 10.1200/JCO.2006.06.209]CENTRAL

Schofield 2013 {published data only}

Schofield P, Ugalde A, Gough K, Reece J, Krishnasamy M, Carey M, et al. A tailored, supportive care intervention using systematic assessment designed for people with inoperable lung cancer: a randomised controlled trial. Psycho‐Oncology 2013;22:2445‐53. [DOI: 10.1002/pon.3306]CENTRAL

Stein 2005 {published data only}

Stein RA, Sharpe L, Bell ML, Boyle FM, Dunn SM, Clarke SJ. Randomized controlled trial of a structured intervention to facilitate end‐of‐life decision making in patients with advanced cancer. Journal of Clinical Oncology 2013;31:3403‐10. [DOI: 10.1200/JCO.2011.40.8872]CENTRAL

Thoonsen 2011 {published data only}

Thoonsen B, Groot M, Engels Y, Prins J, Verhagen S, Galesloot C, et al. Early identification of and proactive palliative care for patients in general practice: incentive and methods of a randomized controlled trial. BMC Family Practice 2011;12:123. [DOI: 10.1186/1471‐2296‐12‐123]CENTRAL
Thoonsen B, Vissers K, Verhagen S, Prins J, Bor H, van Weel C, et al. Training general practitioners in early identification and anticipatory palliative care planning: a randomized controlled trial. BMC Family Practice 2015;16:126. [DOI: 10.1186/s12875‐015‐0342‐6]CENTRAL

Toseland 1995 {published data only}

Toseland RW, Blanchard CG, McCallion P. A problem solving intervention for caregivers of cancer patients. Social Science & Medicine 1995;40:517‐28. [DOI: 10.1016/0277‐9536(94)E0093‐8]CENTRAL

Young 2013 {published data only}

Young JM, Butow PN, Walsh J, Durcinoska I, Dobbins TA, Rodwell L, et al. Multicenter randomized trial of centralized nurse‐led telephone‐based care coordination to improve outcomes after surgical resection for colorectal cancer: the CONNECT intervention. Journal of Clinical Oncology 2013;31:3585‐91. [DOI: 10.1200/JCO.2012.48.1036]CENTRAL

References to studies awaiting assessment

Aljohani 2015 {published data only}

Aljohani A. Early interdisciplinary palliative care for patients with non‐small‐cell lung cancer. 20th Congress of the Asian Pacific Society of Respirology, Kuala Lumpur. (accessed on 25 October 2016); Vol. ID 75:http://www.apsresp.org/congress/apsr2015/oral‐presentations.html. CENTRAL

Groenvold 2017 {published data only}

Groenvold M, Peterson MA, Damkier A, Neergard MA, Nielsen JB, Pedersen L, et al. Randomised clinical trial of early specialist palliative care plus standard care versus standard care alone inpatients with advanced cancer: The Danish Palliative Care Trial. Palliative Medicine 2017;May 1:Online first. [DOI: 10.1177/0269216317705100]CENTRAL
Johnsen AT, Petersen M, Gluud A, Lindschou C, Fayers J, Sjøgren P, et al. Detailed statistical analysis plan for the Danish Palliative Care Trial (DanPaCT). Trials 2014;15:376. [DOI: 10.1186/1745‐6215‐15‐376]CENTRAL
Johnsen AT, Petersen MA, Gluud C, Lindschou J, Fayers P, Sjøgren P, et al. A randomised, multicentre clinical trial of specialised palliative care plus standard treatment versus standard treatment alone for cancer patients with palliative care needs: the Danish palliative care trial (DanPaCT) protocol. BMC Palliative Care 2013;12:37. [DOI: 10.1186/1472‐684X‐12‐37]CENTRAL

Kim 2016 {published data only}

Kim JY, Sun V, Raz DJ, Williams AC, Fujinami R, Reckamp K, et al. The impact of lung cancer surgery on quality of life trajectories inpatients and family caregivers. Lung Cancer 2016;101:35‐9. [DOI: 10.1016/j.lungcan.2016.08.011]CENTRAL

Meyers 2011 {published data only (unpublished sought but not used)}

Meyers FJ, Carducci M, Loscalzo MJ, Linder J, Greasby T, Beckett LA. Effects of a problem‐solving intervention (COPE) on quality of life for patients with advanced cancer on clinical trials and their caregivers: simultaneous care educational intervention (SCEI): linking palliation and clinical trials. Journal of Palliative Medicine 2011;14:465‐73. [DOI: 10.1089/jpm.2010.0416]CENTRAL

NCT00823732 {published data only}

 

NCT01444157 {published data only}

 

NCT02133274 {published data only}

Paiva CE, do Carmo TM, de Oliveira CZ, de Angelis Nascimento MS, De Siquiera MR, Borges M, et al. A phase II randomized study to evaluate a new psychosocial intervention (PI) plus early palliative care (PC) in the reduction of depression of advanced cancer patients (ACP): the PREPArE trial. Journal of Clinical Oncology 2016;34:Suppl; abstr e21626. CENTRAL
do Carmo TM, Paiva BS, de Siqueira MR, da Rosa LT, de Oliveira CZ, Nascimento MS, et al. A phase II study in advanced cancer patients to evaluate the early transition to palliative care (the PREPArE trial): protocol study for a randomized controlled trial. Trials 15;16:160. [DOI: 10.1186/s13063‐015‐0655‐8]CENTRAL

NCT02207322 {published data only}

El‐Jawahri A, LeBlanc TW, Traeger L, VanDusen H, Jackson VA, Greer JA, et al. Randomized trial of an inpatient palliative care intervention in patients hospitalized for hematopoietic stem cell transplantation (HCT). Journal of Clinical Oncology 2016;34:Suppl; abstr 10004. CENTRAL

Temel 2017 {published data only}

Temel JS, El‐Jawahri A, Greer JA, Pirl WF, Jackson VA, Park ER, et al. Randomized trial of early integrated palliative oncology. Journal of Clinical Oncology 2016;34:Suppl; abstr 10003. CENTRAL
Temel JS, Greer JA, El‐Jawahri A, Pirl WF, Park ER, Jackson VA, et al. Effects of early integrated palliative care in patients with lung and GI cancer: a randomized clinical trial. Journal of Clinical Oncology 2017;35:834‐41. [DOI: 10.1200/JCO.2016.70.5046]CENTRAL

Van Arsdale 2016 {published data only}

Van Arsdale AR, Klobocista M, Zanartu C, Pinto P, Rapkin BD, Yi‐Shin Kuo D. Early palliative care intervention for women with gynecologic malignancies. Journal of Clinical Oncology 2016;34:Suppl; abstr e21508. CENTRAL

ACTRN12610000724077 {published data only}

Walczak A, Butow PN, Clayton JM, Tattersall MH, Davidson PM, Young J, et al. Discussing prognosis and end‐of‐life care in the final year of life: a randomised controlled trial of a nurse‐led communication support programme for patients and caregivers. British Medical Journal Open 2014;4:e005745. [DOI: 10.1136/bmjopen‐2014‐005745]CENTRAL

CTRI/2013/11/004128 {published data only}

A study to assess the feasibility of introducing early palliative care in ambulatory patients with advanced lung cancer . Ongoing studySeptember 2013.

CTRI/2016/03/006693 {published data only}

Effect of early integration of specialized palliative care into standard oncologic treatment on the quality of life of patients with advanced head and neck cancers: a randomized controlled trial . Ongoing studyMarch 2016.

DRKS00006162 {published data only}

Meffert C, Gaertner J, Seibel K, Jors K, Bardenheuer H, Buchheidt D, et al. Early Palliative Care‐Health services research and implementation of sustainable changes: the study protocol of the EVI project. BMC Cancer 2015;15:443. [DOI: 10.1186/s12885‐015‐1453‐0]CENTRAL

ISRCTN13337289 {published data only}

Ahmedzai SH, Milroy R, Billingham C, Ryan T, Young T, Gath J, et al. The SPECIAL trial: exploring the place of early specialist palliative care in UK lung cancer management. Lung Cancer 2015;87:S69. [DOI: 10.1016/S0169‐5002(15)50179‐7]CENTRAL

ISRCTN18955704 {published data only}

Gunatilake S, Brims FJ, Fogg C, Lawrie I, Maskell N, Forbes K, et al. A multicentre non‐blinded randomised controlled trial to assess the impact of regular early specialist symptom control treatment on quality of life in malignant mesothelioma (RESPECT‐MESO): study protocol for a randomised controlled trial. Trials 2014;15:367. [DOI: 10.1186/1745‐6215‐15‐367]CENTRAL

NCT01589328 {published data only}

Randomized controlled trials for the effect of early management on PAin and DEpression in patients with PancreatoBiliary cancer (EPADE‐PB) . Ongoing studyApril 2012.

NCT01828775 {published data only}

Integration of palliative care for cancer patients on phase I trials . Ongoing studySeptember 2014.

NCT01865396 {published data only}

Vanbutsele G, Pardon K, Geboes K, De Laat M, Van Belle S, Deliens L. Effect of systematic palliative care on quality of life of patients with advanced cancer of the upper gastrointestinal tract: a randomized controlled trial. Palliative Medicine 2014;28:834‐5. [DOI: 10.1177/0269216314532748]CENTRAL
Vanbutsele G, Van Belle S, De Laat M, Surmont V, Geboes K, Eecloo K, et al. The systematic early integration of palliative care into multidisciplinary oncology care in the hospital setting (IPAC), a randomized controlled trial: the study protocol. BMC Health Services Research 2015;15:554. [DOI: 10.1186/s12913‐015‐1207‐3]CENTRAL

NCT01885884 {published data only}

A pilot trial of an embedded collaborative model of supportive care for pancreatic cancer . Ongoing studyJuly 2013.

NCT01983956 {published data only}

A structured early palliative care intervention for patients with advanced cancer ‐ a randomized controlled trial with a nested qualitative study (SENS trial) . Ongoing studyDecember 2013.

NCT02308865 {published data only}

Impact of early palliative care on quality of life and survival of patients with non‐small‐cell metastatic lung cancer in Northern France . Ongoing studyOctober 2014.

NCT02332317 {published data only}

A randomized, controlled phase III study of integrated, specialized palliative rehabilitation for patients with newly diagnosed non‐resectable cancer . Ongoing studyNovember 2014.

NCT02335619 {published data only}

Early integrated supportive care study for gastrointestinal cancer patients . Ongoing studyFebruary 2015.

NCT02349412 {published data only}

Ongoing studyApril 2015.

NCT02547142 {published data only}

Evaluation of the implementation of an early integrated palliative care program in the esophageal cancer population . Ongoing studyOctober 2015.

NCT02631811 {published data only}

Early palliative care in patients with acute leukaemia (Pablo Hemato) . Ongoing studyNovember 2015.

NCT02712229 {published data only}

A primary palliative care intervention for patients with advanced cancer (CONNECT) . Ongoing studyApril 2016.

NCT02730858 {published data only}

Palliative and oncology care model In breast cancer. Ongoing studyMay 2016.

NCT02853474 {published data only}

Early palliative care in patients with metastatic upper gastrointestinal cancers treated with first‐line chemotherapy (EPIC‐1511) . Ongoing studyAugust 2016.

Addington‐Hall 1995

Addington‐Hall J, McCarthy M. Dying from cancer: results of a national population‐based investigation. Palliative Medicine 1995;9:295‐305. [DOI: 10.1177/026921639500900404]

Alonso‐Coello 2016

Alonso‐Coello P, Oxman AD, Moberg J, Brignardello‐Petersen R, Akl EA, Davoli M, et al. GRADE Evidence to Decision (EtD) frameworks: a systematic and transparent approach to making well informed healthcare choices. 2. Clinical practice guidelines. British Medical Journal 2016;353:i2089. [DOI: 10.1136/bmj.i2089]

American Cancer Society 2013

American Cancer Society. What is advanced cancer?. http://www.cancer.org/treatment/understandingyourdiagnosis/advancedcancer/advanced‐cancer‐what‐is (accessed 31 October 2015).

AUREF 2012

Author and Referee Guide (AUREF), 2012. http://papas.cochrane.org/sites/papas.cochrane.org/files/uploads/L%20‐%20PaPaSAuthor%26RefereeGuidance.pdf (accessed 31 October 2015).

Balshem 2011

Balshem H, Helfand M, Schünemann HJ, Oxman AD, Kunz R, Brozek J, et al. GRADE guidelines: 3. Rating the quality of evidence. Journal of Clinical Epidemiology 2011;64:401‐6. [DOI: 10.1016/j.jclinepi.2010.07.015]

Bauman 2014

Bauman JR, Temel JS. The integration of early palliative care with oncology care: the time has come for a new tradition. Journal of the National Comprehensive Cancer Network 2014;12:1763‐71.

Baumann 2015

Baumann AJ, Wheeler DS, James M, Turner R, Siegel A, Navarro VJ. Benefit of early palliative care intervention in end‐stage liver disease patients awaiting liver transplantation. Journal of Pain and Symptom Management 2015;50:882‐6. [DOI: 10.1016/j.jpainsymman.2015.07.014]

Borenstein 2009

Borenstein M, Hedges LV, Higgins JPT, Rothstein HR. Introduction to Meta‐analysis. Chichester, UK: Wiley, 2009. [DOI: 10.1002/9780470743386]

Boyle 2008

Boyle P, Levin B. World Cancer Report 2008. Lyon, France: International Agency for Research on Cancer, 2008.

Brucker 2005

Brucker PS, Yost K, Cashy J, Webster K, Cella D. General population and cancer patient norms for the Functional Assessment of Cancer Therapy‐General (FACT‐G). Evaluation and the Health Professions 2005;28:192‐211. [DOI: 10.1177/0163278705275341]

Choudhry 2005

Choudhry NK, Fletcher RH, Soumerai SB. Systematic review: the relationship between clinical experience and quality of health care. Annals of Internal Medicine 2005;142:260‐73. [DOI: 10.7326/0003‐4819‐142‐4‐200502150‐00008]

Cohen 1988

Cohen J. Statistical Power Analysis in the Behavioral Sciences. 2nd Edition. Hillsdale, NJ, USA: Lawrence Erlbaum Associates, Inc., 1988.

Davis 2015

Davis MP, Temel JS, Balboni T, Glare P. A review of the trials which examine early integration of outpatient and home palliative care for patients with serious illnesses. Annals of Palliative Medicine 2015;4:99‐121. [DOI: 10.3978/j.issn.2224‐5820.2015.04.04]

de Haes 2005

de Haes H, Teunissen S. Communication in palliative care: a review of recent literature. Current Opinion in Oncology 2005;17:345‐50.

Deeks 2011

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

DerSimonian 1986

DerSimonian R, Laird N. Meta‐analysis in clinical trials. Controlled Clinical Trials 1986;7:177‐88.

Dionne‐Odom 2015

Dionne‐Odom JN, Azuero A, Lyons KD, Hull JG, Tosteson T, Li Z, et al. Benefits of early versus delayed palliative care to informal family caregivers of patients with advanced cancer: outcomes from the ENABLE III randomized controlled trial. Journal of Clinical Oncology 2015;33:1446‐52. [DOI: 10.1200/JCO.2014.58.7824]

Dowsett 2000

Dowsett SM, Saul JL, Butow PN, Dunn SM, Boyer MJ, Findlow R, et al. Communication styles in the cancer consultation: preferences for a patient‐centred approach. Psycho‐Oncology 2000;9:147‐56. [DOI: 10.1002/(SICI)1099‐1611(200003/04)9:2<147::AID‐PON443>3.0.CO;2‐X]

Earle 2008

Earle CC, Landrum MB, Souza JM, Neville BA, Weeks JC, Ayanian JZ. Aggressiveness of cancer care near the end of life: is it a quality‐of‐care issue?. Journal of Clinical Oncology 2008;26:3860‐6. [DOI: 10.1200/JCO.2007.15.8253]

Egger 1997

Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. British Medical Journal 1997;315:629‐34. [DOI: 10.1136/bmj.315.7109.629]

El‐Jawahri 2011

El‐Jawahri A, Greer JA, Temel JS. Does palliative care improve outcomes for patients with incurable illness? A review of the evidence. Journal of Supportive Oncology 2011;9:87‐94. [DOI: 10.1016/j.suponc.2011.03.003]

Elkin 2010

Elkin EB, Bach PB. Cancer's next frontier: addressing high and increasing costs. JAMA 2010;303:1086‐7. [DOI: 10.1001/jama.2010.283]

Ferrell 2017

Ferrell BR, Temel JS, Temin S, Alesi ER, Balboni TA, Basch EM, et al. Integration of palliative care into standard oncology care: American Society of Clinical Oncology clinical practice guideline update. Journal of Clinical Oncology 2017;35:96‐112. [DOI: 10.1200/JCO.2016.70.1474]

Gaertner 2015

Gaertner J, Maier BO, Radbruch L. Resource allocation issues concerning early palliative care. Annals of Palliative Medicine 2015;4:156‐61. [DOI: 10.3978/j.issn.2224‐5820.2015.07.02]

Global Burden of Disease Cancer Collaboration 2015

Global Burden of Disease Cancer Collaboration. The global burden of cancer 2013. JAMA Oncology 2015;1:505‐27. [DOI: 10.1001/jamaoncol.2015.0735]

Gomes 2013

Gomes B, Calanzani N, Curiale V, McCrone P, Higginson IJ. Effectiveness and cost‐effectiveness of home palliative care services for adults with advanced illness and their caregivers. Cochrane Database of Systematic Reviews 2013, Issue 6. [DOI: 10.1002/14651858.CD007760.pub2]

Gotay 2008

Gotay CC, Kawamoto CT, Bottomley A, Efficace F. The prognostic significance of patient‐reported outcomes in cancer clinical trials. Journal of Clinical Oncology 2008;26:1355‐68. [DOI: 10.1200/JCO.2007.13.3439]

GRADEPro GDT 2015 [Computer program]

McMaster University (developed by Evidence Prime, Inc.). GRADEpro Guideline Development Tool [Software]. McMaster University (developed by Evidence Prime, Inc.), 2015.

Greer 2013

Greer JA, Jackson VA, Meier DE, Temel JS. Early integration of palliative care services with standard oncology care for patients with advanced cancer. CA: A Cancer Journal for Clinicians 2013;63:349‐63. [DOI: 10.3322/caac.21192]

Guyatt 2011a

Guyatt GH, Oxman AD, Vist G, Kunz R, Brozek J, Alonso‐Coello P, et al. GRADE guidelines: 4. Rating the quality of evidence ‐ study limitations (risk of bias). Journal of Clinical Epidemiology 2011;64:407‐15. [DOI: 10.1016/j.jclinepi.2010.07.017]

Guyatt 2011b

Guyatt GH, Oxman AD, Kunz R, Woodcock J, Brozek J, Helfand M, et al. GRADE guidelines: 8. Rating the quality of evidence ‐ indirectness. Journal of Clinical Epidemiology 2011;64:1303‐10. [DOI: 10.1016/j.jclinepi.2011.04.014]

Guyatt 2013

Guyatt G, Oxman AD, Sultan S, Brozek J, Glasziou P, Alonso‐Coello P, et al. GRADE guidelines: 11. Making an overall rating of confidence in effect estimates for a single outcome and for all outcomes. Journal of Clinical Epidemiology 2013;66:151‐7. [DOI: 10.1016/j.jclinepi.2012.01.006]

Haun 2014

Haun MW, Sklenarova H, Villalobos M, Thomas M, Brechtel A, Löwe B, et al. Depression, anxiety and disease‐related distress in couples affected by advanced lung cancer. Lung Cancer 2014;86:274‐80. [DOI: 10.1016/j.lungcan.2014.09.009]

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. British Medical Journal 2003;327:557‐60. [DOI: 10.1136/bmj.327.7414.557]

Higgins 2011a

Higgins JPT, Green S. Chapter 4: Guide to the contents of a Cochrane protocol and review. In: Higgins JPT, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions, Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011.

Higgins 2011b

Higgins JPT, Deeks JJ. Chapter 7: Selecting studies and collecting data. In: Higgins JPT, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions, Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011.

Higgins 2011c

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

Higgins 2011d

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

Higgins 2011e

Higgins JP, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, et al. Cochrane Bias Methods Group, Cochrane Statistical Methods Group. The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. British Medical Journal 2011;343:d5928. [DOI: 10.1136/bmj.d5928]

Higginson 2010

Higginson IJ, Evans CJ. What is the evidence that palliative care teams improve outcomes for cancer patients and their families?. Cancer Journal 2010;16:423‐35. [DOI: 10.1097/PPO.0b013e3181f684e5]

Hui 2013a

Hui D, De La Cruz M, Mori M, Parsons HA, Kwon JH, Torres‐Vigil I, et al. Concepts and definitions for "supportive care," "best supportive care," "palliative care," and "hospice care" in the published literature, dictionaries, and textbooks. Supportive Care in Cancer 2013;21:659‐85. [DOI: 10.1007/s00520‐012‐1564‐y]

Hui 2013b

Hui D, Glitza I, Chisholm G, Yennu S, Bruera E. Attrition rates, reasons, and predictive factors in supportive care and palliative oncology clinical trials. Cancer 2013;119:1098‐105. [DOI: 10.1002/cncr.27854]

Hui 2015a

Hui D, Bruera E. Models of integration of oncology and palliative care. Annals of Palliative Medicine 2015;4:89‐98. [DOI: 10.3978/j.issn.2224‐5820.2015.04.01]

Hui 2015b

Hui D, Kim YJ, Park JC, Zhang Y, Strasser F, Cherny N, et al. Integration of oncology and palliative care: a systematic review. Oncologist 2015;1:77‐83. [DOI: 10.1634/theoncologist.2014‐0312]

Hui 2015c

Hui D, Shamieh O, Paiva CE, Perez‐Cruz PE, Kwon JH, Muckaden MA, et al. Minimal clinically important differences in the Edmonton Symptom Assessment Scale in cancer patients: a prospective multicenter study. Cancer 2015;121:3027‐35. [DOI: 10.1002/cncr.29437]

Hussain 2016

Hussain JA, White IR, Langan D, Johnson MJ, Currow DC, Torgerson DJ, et al. Missing data in randomized controlled trials testing palliative interventions pose a significant risk of bias and loss of power: a systematic review and meta‐analyses. Journal of Clinical Epidemiology 2016;74:57‐65. [DOI: 10.1016/j.jclinepi.2015.12.003]

Irwin 2013

Irwin KE, Greer JA, Khatib J, Temel JS, Pirl WF. Early palliative care and metastatic non‐small cell lung cancer: potential mechanisms of prolonged survival. Chronic Respiratory Disease 2013;10:35‐47. [DOI: 10.1177/1479972312471549]

Jacobsen 2011

Jacobsen J, Jackson V, Dahlin C, Greer J, Perez‐Cruz P, Billings JA, et al. Components of early outpatient palliative care consultation in patients with metastatic nonsmall cell lung cancer. Journal of Palliative Medicine 2011;14:459‐64. [DOI: 10.1089/jpm.2010.0382]

Jadad 1996

Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary?. Controlled Clinical Trials 1996;17:1‐12. [DOI: doi.org/10.1016/0197‐2456(95)00134‐4]

Janssens 2015

Janssens A, Teugels L, Kohl S, Michielsen T, van Meerbeeck JP. Integrating early palliative care (EPC) in the management of lung cancer: the role of the thoracic oncologist. Lung Cancer 2015;90:135‐8. [DOI: 10.1016/j.lungcan.2015.08.016]

Janssens 2016

Janssens A, Teugels L, Kohl S, Michielsen T, Leysen B, van Meerbeeck JP. Practical tools for implementing early palliative care in advanced lung cancer. European Respiratory Journal 2016;47:1010‐2. [DOI: 10.1183/13993003.00382‐2015]

Kamal 2016

Kamal AH, LeBlanc TW, Meier DE. Better palliative care for all: improving the lived experience with cancer. JAMA 2016;316:29‐30. [DOI: 10.1001/jama.2016.6491]

Kelley 2010

Kelley AS, Meier DE. Palliative care. A shifting paradigm. New England Journal of Medicine 2010;363:781‐2. [DOI: 10.1056/NEJMe1004139]

Kelley 2015

Kelley AS, Sean Morrison R. Palliative care for the seriously ill. The New England Journal of Medicine 2015;373:747‐55. [DOI: 10.1056/NEJMra1404684]

Lee 2015

Lee RT, Ramchandran K, Sanft T, von Roenn J. Implementation of supportive care and best supportive care interventions in clinical trials enrolling patients with cancer. Annals of Oncology 2015;26:1838‐45. [DOI: 10.1093/annonc/mdv207]

Levy 2016

Levy MH, Smith T, Alvarez‐Perez A, Back A, Baker JN, Beck AC, et al. Palliative care, Version 1.2016. Journal of the National Comprehensive Cancer Network 2016;14:82‐113.

Liberati 2009

Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta‐analyses of studies that evaluate health care interventions: explanation and elaboration. Annals of Internal Medicine 2009;151(4):W65‐94. [DOI: 10.7326/0003‐4819‐151‐4‐200908180‐00136]

Lofgren 2015

Lofgren S, Friedman R, Ghermay R, George M, Pittman JR, Shahane A, et al. Integrating early palliative care for patients with HIV: provider and patient perceptions of symptoms and need for services. American Journal of Hospice and Palliative Care 2015;32:829‐34. [DOI: 10.1177/1049909114550391]

Loudon 2015

Loudon K, Treweek S, Sullivan F, Donnan P, Thorpe KE, Zwarenstein M. The PRECIS‐2 tool: designing trials that are fit for purpose. British Medical Journal 2015;350:h2147. [DOI: 10.1136/bmj.h2147]

Lowery 2013

Lowery WJ, Lowery AW, Barnett JC, Lopez‐Acevedo M, Lee PS, Secord AA, et al. Cost‐effectiveness of early palliative care intervention in recurrent platinum‐resistant ovarian cancer. Gynecologic Oncology 2013;130(3):426‐30. [DOI: 10.1016/j.ygyno.2013.06.011]

Luckett 2014

Luckett T, Phillips J, Agar M, Virdun C, Green A, Davidson PM. Elements of effective palliative care models: a rapid review. BMC Health Services Research 2014;14:136. [DOI: 10.1186/1472‐6963‐14‐136]

May 2015

May P, Garrido MM, Cassel JB, Kelley AS, Meier DE, Normand C, et al. Prospective cohort study of hospital palliative care teams for inpatients with advanced cancer: earlier consultation is associated with larger cost‐saving effect. Journal of Clinical Oncology 2015;33:2745‐52. [DOI: 10.1200/JCO.2014.60.2334]

McClain 2003

McClain CS, Rosenfeld B, Breitbart W. Effect of spiritual well‐being on end‐of‐life despair in terminally‐ill cancer patients. Lancet 2003;361:1603‐7.

McDonald 2016

McDonald J, Swami N, Hannon B, Lo C, Pope A, Oza A, et al. Impact of early palliative care on caregivers of patients with advanced cancer: cluster randomised trial. Annals of Oncology 2016;[Epub ahead of print]:Sep 29. [DOI: 10.1093/annonc/mdw438]

Meffert 2015

Meffert C, Gaertner J, Seibel K, Jors K, Bardenheuer H, Buchheidt D, et al. Early Palliative Care‐Health services research and implementation of sustainable changes: the study protocol of the EVI project. BMC Cancer 2015;15:443. [DOI: 10.1186/s12885‐015‐1453‐0]

Mehnert 2014

Mehnert A, Brähler E, Faller H, Härter M, Keller M, Schulz H, et al. Four‐week prevalence of mental disorders in patients with cancer across major tumor entities. Journal of Clinical Oncology 2014;32:3540‐6. [DOI: 10.1200/JCO.2014.56.0086]

Meyers 2003

Meyers FJ, Linder J. Simultaneous care: disease treatment and palliative care throughout illness. Journal of Clinical Oncology 2003;21:1412‐5.

Morrison 2004

Morrison RS, Meier DE. Clinical practice. Palliative care. New England Journal of Medicine 2004;350:2582‐90. [DOI: 10.1056/NEJMcp035232]

Movsisyan 2016a

Movsisyan A, Melendez‐Torres GJ, Montgomery P. Users identified challenges in applying GRADE to complex interventions and suggested an extension to GRADE. Journal of Clinical Epidemiology 2016;70:191‐9. [DOI: 10.1016/j.jclinepi.2015.09.010]

Movsisyan 2016b

Movsisyan A, Melendez‐Torres GJ, Montgomery P. Outcomes in systematic reviews of complex interventions never reached “high” GRADE ratings when compared with those of simple interventions. Journal of Clinical Epidemiology 2016;78:22‐33. [DOI: 10.1016/j.jclinepi.2016.03.014]

Nipp 2016

Nipp RD, Greer JA, El‐Jawahri A, Traeger L, Gallagher ER, Park ER, et al. Age and gender moderate the impact of early palliative care in metastatic non‐small cell lung cancer. The Oncologist 2016;21:119‐26. [DOI: 10.1634/theoncologist.2015‐0232]

Parikh 2013

Parikh RB, Kirch RA, Smith TJ, Temel JS. Early specialty palliative care ‐ translating data in oncology into practice. New England Journal of Medicine 2013;369:2347‐51. [DOI: 10.1056/NEJMsb1305469]

Peppercorn 2011

Peppercorn JM, Smith TJ, Helft PR, Debono DJ, Berry SR, Wollins DS, et al. American Society of Clinical Oncology statement: toward individualized care for patients with advanced cancer. Journal of Clinical Oncology 2011;29:755‐60. [DOI: 10.1200/JCO.2010.33.1744]

Piggott 2004

Piggott M, McGee H, Feuer D. Has CONSORT improved the reporting of randomized controlled trials in the palliative care literature? A systematic review. Palliative Medicine 2004;18:32‐8. [DOI: 10.1191/0269216304pm857oa]

Pinquart 2010

Pinquart M, Duberstein PR. Depression and cancer mortality: a meta‐analysis. Psychological Medicine 2010;40:1797‐810. [DOI: 10.1017/S0033291709992285]

Prigerson 2015

Prigerson HG, Bao Y, Shah MA, Paulk ME, LeBlanc TW, Schneider BJ, et al. Chemotherapy use, performance status, and quality of life at the end of life. JAMA Oncology 2015;1:778‐84. [DOI: 10.1001/jamaoncol.2015.2378]

Quaresma 2015

Quaresma M, Coleman MP, Rachet B. 40‐Year trends in an index of survival for all cancers combined and survival adjusted for age and sex for each cancer in England and Wales, 1971–2011: a population‐based study. The Lancet 2015;385:1206‐18. [DOI: 10.1016/S0140‐6736(14)61396‐9]

RevMan 2014 [Computer program]

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

Roberts 2015

Roberts I, Ker K. How systematic reviews cause research waste. The Lancet 2015;386:1536.

Salins 2016

Salins N, Ramanjulu R, Patra L, Deodhar J, Muckaden MA. Integration of early specialist palliative care in cancer care and patient related outcomes: a critical review of evidence. Indian Journal of Palliative Care 2016;22:253‐7. [DOI: 10.4103/0973‐1075.185028]

Schenker 2015

Schenker Y, Arnold R. The next era of palliative care. JAMA 2015;314:1565‐6. [DOI: 10.1001/jama.2015.1121]

Schünemann 2011

Schünemann HJ, Oxman AD, Vist GE, Higgins JPT, Deeks JJ, Glasziou P, et al. Chapter 12: Interpreting results and drawing conclusions. In: Higgins JPT, Green S editor(s). Cochrane Handbook for Systematic Reviews of Interventions, Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011.

Simone 2nd 2015

Simone CB. Early palliative care and integration of palliative care models in modern oncology practices. Annals of Palliative Medicine 2015;4:84‐6. [DOI: 10.3978/j.issn.2224‐5820.2015.07.07]

Sinclair 2006

Sinclair CT. Communicating a prognosis in advanced cancer. Journal of Supportive Oncology 2006;4:201‐4.

Singer 1999

Singer PA, Martin DK, Kelner M. Quality end‐of‐life care: patients' perspectives. JAMA 1999;281:163‐8. [DOI: 10.1001/jama.281.2.163]

Sklenarova 2015

Sklenarova H, Krümpelmann A, Haun MW, Friederich HC, Huber J, Thomas M, et al. When do we need to care about the caregiver? Supportive care needs, anxiety, and depression among informal caregivers of patients with cancer and cancer survivors. Cancer 2015;121:1513‐9. [DOI: 10.1002/cncr.29223]

Smith 2003

Smith TJ, Coyne P, Cassel B, Penberthy L, Hopson A, Hager MA. A high‐volume specialist palliative care unit and team may reduce in‐hospital end‐of‐life care costs. Journal of Palliative Medicine 2003;6:600‐705. [DOI: 10.1089/109662103322515202]

Smith 2012

Smith TJ, Temin S, Alesi ER, Abernethy AP, Balboni TA, Basch EM, et al. American Society of Clinical Oncology provisional clinical opinion: the integration of palliative care into standard oncology care. Journal of Clinical Oncology 2012;30:880‐7. [DOI: 10.1200/JCO.2011.38.5161]

Sterne 2011

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

Tassinari 2016

Tassinari D, Drudi F, Monterubbianesi MC, Stocchi L, Ferioli I, Marzaloni A, et al. Early palliative care in advanced oncologic and non‐oncologic chronic diseases. A systematic review of the literature. Reviews on Recent Clinical Trials 2016;11:1. [DOI: 10.2174/1574887110666151014141650]

Thompson 2002

Higgins J, Thompson S, Deeks J, Altman D. Statistical heterogeneity in systematic reviews of clinical trials: a critical appraisal of guidelines and practice. Journal of Health Services Research & Policy 2002;7:51‐61. [DOI: 10.1258/1355819021927674]

Thorpe 2009

Thorpe KE, Zwarenstein M, Oxman AD, Treweek S, Furberg CD, Altman DG, et al. A pragmatic‐explanatory continuum indicator summary (PRECIS): a tool to help trial designers. Journal of Clinical Epidemiology 2009;5:464‐75. [DOI: 10.1016/j.jclinepi.2008.12.011]

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. [DOI: 10.1186/1745‐6215‐8‐16]

Trajkovic‐Vidakovic 2012

Trajkovic‐Vidakovic M, de Graeff A, Voest EE, Teunissen SCCM. Symptoms tell it all: a systematic review of the value of symptom assessment to predict survival in advanced cancer patients. Critical Reviews in Oncology/Hematology 2012;84:130‐48. [DOI: 10.1016/j.critrevonc.2012.02.011]

Treweek 2009

Treweek S, Zwarenstein M. Making trials matter: pragmatic and explanatory trials and the problem of applicability. Trials 2009;10:37. [DOI: 10.1186/1745‐6215‐10‐37]

van Mechelen 2013

van Mechelen W, Aertgeerts B, De Ceulaer K, Thoonsen B, Vermandere M, Warmenhoven F, et al. Defining the palliative care patient: a systematic review. Palliative Medicine 2013;27:197‐208. [DOI: 10.1177/0269216311435268]

von Roenn 2011

von Roenn JH, Temel J. The integration of palliative care and oncology: the evidence. Oncology (Williston Park) 2011;25:1266.

Weber 2014

Weber C, Stirnemann J, Herrmann FR, Pautex S, Janssens JP. Can early introduction of specialized palliative care limit intensive care, emergency and hospital admissions in patients with severe and very severe COPD? A randomized study. BMC Palliative Care 2014;13:47. [DOI: 10.1186/1472‐684X‐13‐47]

Wee 2008

Wee B, Hadley G, Derry S. How useful are systematic reviews for informing palliative care practice? Survey of 25 Cochrane systematic reviews. BMC Palliative Care 2008;7:13. [DOI: 10.1186/1472‐684X‐7‐13]

WHO 2013

World Health Organization. Definition of palliative care. http://www.who.int/cancer/palliative/definition/en/ (accessed 31 October 2015).

Wolfson 2015

Wolfson JA, Sun CL, Wyatt LP, Hurria A, Bhatia S. Impact of care at comprehensive cancer centers on outcome: results from a population‐based study. Cancer 2015;121:3885‐93. [DOI: 10.1002/cncr.29576]

Yoong 2013

Yoong J, Park ER, Greer JA, Jackson VA, Gallagher ER, Pirl WF, et al. Early palliative care in advanced lung cancer: a qualitative study. JAMA Internal Medicine 2013;173:283‐90. [DOI: 10.1001/jamainternmed.2013.1874]

Zambrano 2016

Zambrano SC, Fliedner MC, Eychmüller S. The impact of early palliative care on the quality of care during the last days of life: what does the evidence say?. Current Opinion in Supportive and Palliative Care 2016;10:310‐5. [DOI: 10.1097/SPC.0000000000000240]

Zhang 2009

Zhang B, Wright AA, Huskamp HA, Nilsson ME, Maciejewski ML, Earle CC, et al. Health care costs in the last week of life: associations with end of life conversations. Archives of Internal Medicine 2009;169:480‐8. [DOI: 10.1001/archinternmed.2008.587]

Zhi 2015

Zhi WI, Smith TJ. Early integration of palliative care into oncology: evidence, challenges and barriers. Annals of Palliative Medicine 2015;4:122‐31. [DOI: 10.3978/j.issn.2224‐5820.2015.07.03]

Zimmermann 2008

Zimmermann C, Riechelmann R, Krzyzanowska M, Rodin G, Tannock I. Effectiveness of specialized palliative care: a systematic review. JAMA 2008;299:1698‐709. [DOI: 10.1001/jama.299.14.1698]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Bakitas 2009

Methods

Parallel‐group randomised controlled trial (RCT)

Participants

Country and regions: USA, rural area; Norris Cotton Cancer Center (NCCC) and Dartmouth College in Lebanon, New Hampshire, and affiliated outreach clinics, Veterans Administration Medical Center (VAMC), in White River Junction, Vermont

Recruitment: identification by research assistants at NCCC and VAMC tumour boards (gastrointestinal, genitourinary, breast, and thoracic cancer management meetings) and review of clinicians' clinic schedules

Inclusion criteria: 18 years of age or older; life‐limiting cancer (prognosis approximately 1 year); and within 8 to 12 weeks of a new diagnosis of gastrointestinal tract (unresectable stage III or IV), lung (stage IIIB or IV non–small cell or extensive small cell), genitourinary tract (stage IV, prostate cancers limited to persons with hormone refractory), or breast (stage IV and visceral crisis, lung or liver metastasis, oestrogen receptor (ER) negative, human epidermal growth factor receptor 2 (Her 2 neu) positive)) cancer

Exclusion criteria: impaired cognition (< 17 on the Adult Lifestyles and Function Interview‐Mini Mental State Exam); and Axis I psychiatric disorder (schizophrenia, bipolar disorder) or active substance use disorder

Number of participants enrolled (survival outcomes sample): N = 322 (161 intervention and 161 control) randomised

Participant characteristics (patient outcomes sample): N = 279 (87% returned baseline questionnaires); mean age (intervention/control in years): 65.4/65.2; male gender (intervention/control in %): 62.1/58.2; married or living with partner (intervention/control in %): 73.1/67.2; education < 9 years (intervention/control in %): 11.7/14.9; Caucasian (intervention/control in %): 98.6/98.5; employed (intervention/control in %): 20.0/16.4; live in rural setting (intervention/control in %): 52.4/60.5; Karnofsky Performance Status (intervention/control mean): 78.4/77.4; differences between intervention and control not statistically significant
Diseases (patient outcomes sample, intervention/control in %): gastrointestinal tract cancer 42.1/43.3; lung cancer 34.5/32.1; genitourinary 13.1/13.4; breast 10.3/11.2; differences between intervention and control not statistically significant

Deaths at end of study (intervention/control in N (%)): 112 (69.6)/119 (73.9); differences between intervention and control not statistically significant
Withdrawals/other drop‐outs (intervention/control in N (%)): 16 (9.9)/27 (16.8); differences between intervention and control not statistically significant (P = 0.10)

Number of caregivers enrolled (intervention/control in N): 116/104
Caregiver characteristics (caregiver outcomes sample): mean age 59.0 years; male 22.7%; married or living with partner 83.3%; education < 9 years 6.1%; Caucasian 96.5%; employed 42.9%; relationship to patient: spouse/partner 70.7%, child 16.2%

Interventions

Name: educational and care management palliative care intervention for persons with advanced cancer and a caregiver compared with care as usual (project ENABLE II)

Service base: outpatient palliative care

Intervention condition (n = 161): case management and educational approach with a manualised, telephone‐based format carried out by 2 advanced practice nurses with palliative care specialty training; 4 initial structured educational and problem‐solving telephone sessions on a weekly basis (education manual: Charting your Course: An Intervention for People and Families Living With Cancer) and at least monthly telephone follow‐up sessions thereafter until the participant died or the study ended; problem‐solving management on the basis of systematic distress assessment using the Distress Thermometer with a cut‐off > 3; when concerns were identified, participants were encouraged to contact the oncology or palliative care clinical teams; monthly medical appointments for participants and their caregivers (attendance in person or by toll‐free conference call) led by a palliative care physician and nurse practitioner, biweekly study team meetings to review audiotaped educational sessions with regards to difficult patient management issues

Control condition (n = 161): free access to all oncology and supportive services without restrictions, including referral to the institution's interdisciplinary palliative care service for symptom and supportive care, free access to an advanced illness co‐ordinated care program (Advanced Illness Care Committee, AICC) that provided consultation to oncology staff for inpatients with life‐limiting illness at the VAMC (including prognosis and goals of care assessment, pain and symptom management, advance care planning, referral to hospice)

Outcomes

Primary endpoints: patient‐reported quality of life (Functional Assessment of Chronic Illness Therapy‐Palliative Care, FACIT‐Pal), symptom intensity (Edmonton Symptom Assessment Scale, ESAS), and resource use (number of days in the hospital, number of days in intensive care unit, and number of emergency department visits)

Secondary endpoints: mood status (Center for Epidemiological Studies‐Depression scale, CES‐D), survival, caregiver burden (Montgomery Borgatta Caregiver Burden Scale), quality of care (After Death Bereaved Family Member Interview, ADI)

Assessment points: baseline/T0: after randomisation; T1: 1 month after baseline; then every 3 months until the participant died or the study was completed (31 December 2007)

Notes

Funding source: National Cancer Institute (NCI), USA

Declarations of interest among primary researchers: no financial disclosures reported. Dr Bakitas was a recipient of a Department of Defense Clinical Nurse Researcher award, an American Cancer Society Doctoral scholarship, and a postdoctoral fellowship at Yale University School of Nursing (National Institutes of Health/National Institute of Nursing Research grant T32NR008346). This study was supported by National Cancer Institute grant R01 CA101704

Power considerations: At study completion, final enrolment was 322 owing to slower accrual than was projected in the initial power calculation (target sample size of 400, 80% power for scores on FACIT‐Pal, ESAS, and CES‐D based on a t test comparing treatment groups with respect to the last observed value at a 2‐sided alpha of .01). Reduced sample size and power might have increased the probability of type II error

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from main publication: "Participants were randomised equally into either the intervention or the usual care group using computer‐generated random numbers"

Judgement: probably done, as investigators consistently describe the use of random sequences

Allocation concealment (selection bias)

High risk

Quote from main publication: "Referring clinicians were neither informed nor formally blinded to participant assignment"

Judgement: probably not done

Blinding of participants (performance bias)

High risk

Preregistration on clinicaltrials.gov (NCT00253383); says it was an open‐label trial

Judgement: not done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Original publication does not explicitly address blinding of outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Judgement: N = 113 completers in intervention group vs N = 105 completers in control group (Fisher's exact test with 2‐tailed P = 0.40)

Selective reporting (reporting bias)

Low risk

Judgement: All outcomes from clinicaltrials.gov registration listed and reported in publications

Other bias

Low risk

None detected

Bakitas 2015

Methods

Parallel‐group randomised controlled trial (RCT) with fast‐track/delayed‐intervention design

Participants

Country and regions: USA, Norris Cotton Cancer Center and Dartmouth College in Lebanon, New Hampshire, Veterans Administration (VA) medical centre in White River Junction, Vermont

Recruitment: 29 months, identification by research assistants/research co‐ordinators reviewing all outpatient clinicians’ schedules and tumour board lists

Inclusion criteria: able to speak and understand English; over the age of 18; new diagnosis, recurrence, or progression of advanced stage cancer within approximately 60 days of the date the patient was informed of the diagnosis by his/her oncology clinician; estimated survival of 2 years or less; diagnosed advanced stage solid tumour such as one of the following:

• lung cancer: stage IIIB or IV non‐small cell, or extensive stage small cell

• breast cancer: stage IV with poor prognostic indicators including but not limited to > 2 cytotoxic regimens for metastatic breast cancer (MBC) or diagnosis of MBC ≤ 12 months since completion of adjuvant or neo‐adjuvant treatment, or triple negative disease (ER/PR and HER‐) or parenchymal brain mets and/or carcinomatous meningitis

gastrointestinal cancers: unresectable stage III or IV

• genitourinary cancers: stage IV (for prostate cancer inclusion is limited to persons with hormone refractory prostate cancer)

• brain cancer: unresectable; grade IV

• melanoma: stage IV

• haematological malignancies: leukaemia (e.g. acute myeloid leukaemia, acute lymphoblastic leukaemia, chronic myeloid leukaemia, chronic lymphoblastic leukaemia) with advanced stage, treatment refractory, poor prognosis cell type or chromosomal abnormalities, “older age”; lymphoma with stage IV, treatment‐refractory Hodgkin’s disease or non‐Hodgkin’s lymphoma; multiple myeloma with elevated β2‐microglobulin, albumin < 3.5 g/dL, plasma cell labelling index > 1%, CRP > 6 µg/mL, elevated lactate dehydrogenase, plasmablastic morphology, abnormal chromosome 13

Exclusion criteria: dementia or significant confusion (impaired cognitive status as indicated by a score ≤ 3 on the Callahan 6‐item cognitive screening tool); Axis I psychiatric diagnosis of severe mental illness (DSM‐IV) (e.g. schizophrenia, bipolar disorder, active substance use disorder); patients were not excluded if they had not identified a caregiver; 4. prior involvement with palliative care service within the past year; minimum predicted survival < 12 weeks (3 months)

Number of participants enrolled: N = 207 (104 intervention and 103 control) randomised

Patient characteristics: N = 207; mean age (intervention/control in years): 64.0/64.6; male gender (intervention/control in %): 53.9/51.5; married or living with partner (intervention/control in %): 73.1/69.7; education < 9 years (intervention/control in %): 7.7/2.9; Caucasian (intervention/control in %): 98.1/95.2; employed (intervention/control in %): 24.0/23.3; live in rural setting (intervention/control in %): 59.6/58.3; Karnofsky Performance Status (intervention/control mean): 80.6/81.5; intervention group with significantly less education, higher weekly alcoholic beverage use, and higher clinical trial enrolment
Diseases (intervention/control in %): lung cancer 44.2/40.8; gastrointestinal tract 25.0/23.3; breast 9.6/12.6; other solid tumour 9.6/9.7; genitourinary tract 6.7/8.7; haematological malignancy 4.5/4.6; differences between intervention and control not statistically significant

Deaths at end of study (intervention/control in N (%)): 50 (48.1)/59 (57.3); difference between intervention and control not statistically significant
Withdrawals/other drop‐outs (intervention/control in N(%)): 12 (11.5)/22 (21.4); difference between intervention and control not statistically significant (P = 0.06)

Number of caregivers enrolled (intervention/control in N): 63/61
Caregiver characteristics (intervention/control in N (%)): mean age (intervention/control in years): 61/57.9; male gender (intervention/control in %): 23/19.7; married or living with partner (intervention/control in %): 88.5./95.1; education < 9 years (intervention/control in %): 0/1.6; Caucasian (intervention/control in %): 90.2/95.1; employed (intervention/control in %): 37.7/23.3; relationship to patient (intervention/control in %): spouse/partner 78.7/72.1, child 6.6/16.4, sibling 4.9/6.6, parent 6.6/4.9

Interventions

Name: early vs. later palliative cancer care: clinical and biobehavioural outcomes (project ENABLE III). All participants received usual oncology care directed by a medical oncologist and consisting of anticancer and symptom control treatments and consultation with oncology and supportive care specialists, including a clinical palliative care team, which was provided whenever requested, regardless of group assignment

Service base: outpatient palliative care

Intervention condition (n = 104): ENABLE telehealth concurrent palliative care model within 30 to 60 days of being informed of an advancer cancer diagnosis, cancer recurrence, or progression: initial in‐person, standardised outpatient palliative care consultation by a board‐certified palliative care clinician and 6 structured weekly telephone coaching sessions by an advanced practice nurse using a manualised curriculum (Charting Your Course: An Intervention for Patients With Advanced Cancer); sessions 1 to 3 focussed on problem solving, symptom management, self‐care, identification and co‐ordination of local resources, communication, decision making, and advance care planning; sessions 4 to 6 comprised Outlook, a life‐review approach that encourages participants to frame advanced illness challenges as personal growth opportunities; after the 6 Charting Your Course sessions, monthly follow‐up calls reinforced prior content and identified new challenges or care co‐ordination issues; study principal investigator reviewed all palliative care consultation notes and digitally recorded nurse coach sessions for protocol adherence; principal investigator also met with nurse coaches weekly to review and provide feedback on difficult cases

Control condition (n = 103): ENABLE telehealth concurrent palliative care model 3 months after being informed of an advancer cancer diagnosis, cancer recurrence, or progression

Outcomes

Primary endpoints: patient‐reported quality of life (FACIT‐Pal) and Trial Outcome Index (TOI), symptom impact (QUAL‐E), mood (CES‐D), 1‐year and overall survival, resource use (patient‐reported hospital and intensive care unit days and emergency department visits, decedents' data for the period between last patient‐reported assessment and death, chemotherapy use in last 14 days and location of death via medical record review)

Secondary endpoints: caregiver burden, location of death

Assessment points: baseline/T0: before randomisation; T1: 3 months from enrolment; T2: 6 months from enrolment; T3: 12 months from enrolment; in terminal decline joint modelling: T1: 12 months before death; T2: 6 months before death; T3: 3 months before death

Notes

Funding source: grant no. R01NR011871‐01 from the National Institute for Nursing Research; Cancer and Leukemia Group B Foundation Clinical Scholar Award; Foundation for
Informed Medical Decision‐Making; by grant nos. P30CA023108, UL1 TR001086, and R03NR014915;
NIH/NINR Small Research Grant 1R03NR014915‐01 (Zhigang Li); Norris Cotton Cancer Center pilot funding; Dartmouth‐Hitchcock Section of Palliative Medicine; National Palliative Care Research Center Junior Career Development Award (M.A.B.); grant no. 5R25CA047888 from the University of Alabama at Birmingham Cancer Prevention and Control Training Program (J.N.D.‐O.); Mentored Research Scholar grant no. MRSG 12‐113‐01‐CPPB in Applied and Clinical Research from the American Cancer Society (K.D.L.)

Declarations of interest among primary researchers: Mark T. Hegel reported research funding from Johnson & Johnson. Remaining study authors reported no relationships to disclose

Power considerations: At planned study completion date (15 March 2013), final enrolment was 207 because of slower than anticipated accrual. On the basis of final sample size, 3‐month detectable differences were 7.7 points for FACIT‐Pal and 3.2 points for CES‐D and thus was larger than projected in the initial power calculation (target sample size of 360, 80% power to detect a 6‐point difference in FACIT‐Pal and 2.5‐point difference in CES‐D based on a t test comparing 3‐month group differences at a 2‐sided alpha of .05). Reduced sample size and power might have impeded detection of differences (type II error) in patient‐reported outcomes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Random assignment was on a one‐to‐one basis using computer‐generated randomly permuted treatment assignments with randomly assigned block sizes of two and four stratified by disease (six categories) and enrolment site (four clinics)"

Judgement: probably done, as investigators consistently describe the use of random sequences

Allocation concealment (selection bias)

Unclear risk

Original publication does not explicitly address allocation concealment

Judgement: unclear risk of bias

Blinding of participants (performance bias)

High risk

Preregistration on clinicaltrials.gov (NCT01245621) says it was an open‐label trial that blinded only outcome assessors

Judgement: not done

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quote from main publication: "Data collectors were blinded to participant group"

Judgement: probably done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Judgement: N = 59 completers in intervention group vs N = 54 completers in control group (Fisher's exact test with 2‐tailed P = 0.58)

Selective reporting (reporting bias)

Low risk

Judgement: Inventory of Complicated Grief‐Short Form (ICG‐SF) and Quality of Death and Dying (QODD) as outcomes from clinicaltrials.gov registration not reported in publications so far. Publication does not differentiate between primary and secondary outcomes. However, all key outcomes have been reported. We made a close‐call decision favouring low risk at this point against unclear risk of bias

Other bias

Low risk

None detected

Maltoni 2016

Methods

Parallel group cluster‐randomised controlled trial (cRCT)

Participants

Country and regions: Italy, 21 centres

Recruitment: 29 months, patients with advanced and/or metastatic pancreatic cancer

Inclusion criteria: diagnosis of inoperable locally advanced and/or metastatic pancreatic; cancer for a maximum of 8 weeks before enrolment; age ≥ 18 years; Eastern Cooperative Oncology Group
(ECOG) performance status 0‐2; life expectancy > 2 months; candidate for antitumour treatment (chemotherapy or target therapy); newly referred patients

Exclusion criteria: patients who were already receiving PC; patients who had received prior chemotherapy for metastatic or advanced disease; patients who had participated in a clinical trial

Number of participants enrolled: N = 186 (89 intervention and 97 control)

Participant characteristics: N = 186; median age (intervention/control in years): 66/67; male gender (intervention/control in %): 61.5/52.8; married or living with partner (intervention/control in %): 76.9/78.6; ECOG performance status 0, 1, 2 (intervention/control in %): 56.7, 37.1, 6.2/56.2, 39.3, 4.5; differences between intervention and control not statistically significant with respect to age, martial status, and performance status

Diseases (intervention/control in %): metastatic pancreatic cancer 100/100

Deaths at end of study (intervention/control in N (%)): 19 (19.6)/16 (17.8); differences between intervention and control not statistically significant

Withdrawals/other drop‐outs (intervention/control in N): 33/24; differences between intervention and control not statistically significant

Interventions

Name: standard cancer care plus on‐demand early palliative care or standard cancer care plus systematic early palliative care (interventional arm)

Service base: 21 Italian centres

Intervention condition (n = 89): "Patients assigned to the interventional arm had an appointment scheduled with a PC specialist who had a predefined checklist of issues to be addressed during the consultation. The use of the checklist by the individual researcher was not monitored from the outside, but reported by the researcher himself. The checklist of topics to be discussed during the visit of PC is the same used by Temel [4] and is reported in the original protocol. Patients met a member of the PC team within 2 weeks of enrolment and were seen thereafter every 2 to 4 weeks until death. In both arms, availability between appointments not scheduled in the protocol, but according to the clinical and organisational solutions, was present in every centre. Moreover, every researcher could have adjunctive routine tools of assessment, not considered in the present study. Palliative care appointments and interventions were oriented by general PC guidelines [12]. The full‐time palliative care specialist who regularly saw interventional arm patients could prescribe drugs and request other interventions pertaining to physical, psychological, and spiritual needs. However, recommendations made by the PC expert on decision making processes had to be shared by the oncologist"

Control condition (n = 97): "Patients assigned to the standard arm were not scheduled to meet the PC team unless they, their families, or the attending oncologist requested an appointment. After the evaluation period (T1 = 12 +/‐3 weeks from T0), patients were followed by the PC team as needed"

Outcomes

Primary endpoints: health‐related quality of life (Trial Outcome Index, TOI, as sum of scores on the disease‐specific subscale and on physical and functional well‐being subscales of the Functional Assessment of Cancer Therapy‐Hepatobiliary, FACT‐Hep)

Secondary endpoints: mood (Hospital Anxiety and Depression Scale, HADS), family satisfaction with end‐of‐life care (FAMCARE), end‐of‐life care aggressiveness (chemotherapy in the last 30 days of life, median duration of hospice admission, death at home or in hospice)

Assessment points: baseline/T0: before randomisation; T1: 12 +‐/+ 3 weeks from enrolment

Notes

Funding source: grant no. RF‐2011‐02350971 from the Italian Ministry of Health

Declarations of interest among primary researchers: Study authors declared no conflicts of interest

Power considerations: At study completion, final enrolment was 186 and was somewhat lower than projected in the initial power calculation (target sample size of 240, 80% power on a t test comparing treatment groups at a 2‐sided alpha of .05, effect size 0.50). Reduced sample size and power might have increased probability of type II error

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "Eligible patients were randomised for a maximum of 8 weeks after diagnosis and before anticancer treatment to one of the two groups on a 1:1 allocation rate. Separate randomisation lists using a permuted block balanced procedure were generated for each participating centre"

Judgement: probably done

Allocation concealment (selection bias)

Low risk

Quote (reply received from principal investigator): "The random assignment was done by a telephone call to the Biostatistics and Clinical Trials Unit of the coordinating center in Meldola using computer‐generated randomization lists of permutated blocks of varying sizes stratified for participating center. The sequences were concealed from the physicians" Judgement: probably done

Blinding of participants (performance bias)

High risk

Quote: "No masking was involved in this open‐label trial" Judgement: not done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Original publication does not explicitly address blinding of outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Judgement: N = 64 completers in intervention group vs N = 65 completers in control group (Fisher's exact test with 2‐tailed P value at 0.34)

Selective reporting (reporting bias)

Low risk

Judgement: all outcomes from clinicaltrials.gov registration listed and reported in publications

Other bias

Low risk

None detected

McCorkle 2015

Methods

Parallel‐group cluster‐randomised controlled trial (cRCT)

Participants

Country and regions: USA, Smilow Cancer Hospital at Yale/New Haven, Connecticut

Recruitment: 29 months, gynaecological, lung, head and neck, and gastrointestinal clinics, patients identified at weekly tumour boards and approached by their oncologist

Inclusion criteria: aged 21 years or older; late‐stage cancer diagnosis within 100 days; post biopsy or surgery with additional treatment recommended; at least 1 self‐reported chronic condition

Exclusion criteria: not reported

Number of participants enrolled: N = 146 (66 intervention and 80 control), gynaecological and lung clinics allocated to intervention group, head and neck and gastrointestinal clinics allocated to control group

Participant characteristics: N = 146; age < 65 years (intervention/control in %): 51.5/71.3; age ≥ 65 years (intervention/control in %): 48.5/28.7; male gender (intervention/control in %): 28.8/56.3; married or living with partner (intervention/control in %): 60.6/52.5; education < 9 years (intervention/control in %): 27.3/30.3; employed (intervention/control in %): 30.3/37.5; number of comorbidities between 3 and 12 (intervention/control in %): 63.6/36.2; differences between intervention and control statistically significant with respect to age, gender, and comorbidity

Diseases (intervention/control in %): lung cancer 25.3/‐‐; gastrointestinal tract ‐‐/36.3; gynaecological tumour 19.9/‐‐; head and neck tumour 0/18.5

Deaths at end of study (intervention/control in N (%)): 7 (10.6)/3 (3.8); differences between intervention and control not statistically significant

Withdrawals/other drop‐outs (intervention/control in N (%)): 23 (34.8)/21 (26.2); differences between intervention and control not statistically significant

Interventions

Name: advanced practice nurse co‐ordinated multi‐disciplinary intervention vs standard cancer care

Service base: 4 disease‐specific outpatient clinics

Intervention condition (n = 66): 10‐week standardised intervention delivered by different members of each team included monitoring participants' status, providing symptom management, executing complex care procedures, teaching participants and family caregivers, clarifying the illness experience, co‐ordinating care, responding to the family, enhancing quality of life, and collaborating with other providers; clinic advanced practice nurses initially contacted participants within 24 hours, and weekly phone and scheduled in‐person contacts (5 clinic visits and 5 telephone calls); members of each disease‐specific multi‐disciplinary team worked together as a palliative care unit, each member taking on different functions to ensure all components of the intervention were addressed; clinic advanced practice nurse oversaw co‐ordination and implementation

Control condition (n = 80): enhanced usual care, i.e. usual multi‐disciplinary care plus a copy of the symptom management toolkit with instructions on its use

Outcomes

Primary endpoints: symptom distress (Symptom Distress Scale, SDS), health distress (4‐item scale developed by the Stanford Patient Education Research Center), depression (Patient Health Questionnaire, PHQ‐9), emotional distress (Emotional Distress Thermometer, EDT), functional status (Enforced Social Dependency Scale, ESDS), self‐rated health (first item of the SF‐12)

Secondary endpoints: anxiety (7‐item Hospital Anxiety and Depression Scale, HADS‐Anxiety), self‐efficacy (Self‐Efficacy for Managing Chronic Disease Scale, SEMCD), uncertainty (Mishel Uncertainty in Illness Scale ‐ Community Form, MUIS‐C), quality of life (Functional Assessment of Cancer Therapy ‐ General, FACT‐G)

Assessment points: baseline/T0: after randomisation; T1: 1 month from enrolment; T2: 3 months from enrolment

Notes

Funding source: NIH/NINR grant R01NR011872

Declarations of interest among primary researchers: Apart from funding, no further study author disclosure statements were made

Power considerations: No a priori sample size calculation was provided. Small sample size and power might have increased probability of type II error

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from main publication: "Randomization was done using the ranuni function in conjunction with the rank procedure in statistical software SAS (SAS version 9.2 for Windows; SAS Institute Inc., Cary, NC)" Judgement: probably done

Allocation concealment (selection bias)

Unclear risk

Original publication does not explicitly address allocation concealment. Judgement: unclear risk of bias

Blinding of participants (performance bias)

High risk

Preregistration on clinicaltrials.gov (NCT01272024) says it was an open‐label trial. Judgement: unclear risk of bias

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Original publication does not explicitly address blinding of outcome assessment. Judgement: unclear risk of bias

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Judgement: N = 36 completers in intervention group vs N = 56 completers in control group (Fisher's exact test with 2‐tailed P = 0.06). Non‐significant trend for higher attrition in the intervention group. We made a close‐call decision favouring low risk against high risk of bias

Selective reporting (reporting bias)

High risk

Uncertainty (MUIS‐C) as single primary outcome in clinicaltrials.gov registration, reported as secondary outcome in publication. Anxiety (HADS‐Anxiety) as single secondary outcome in clinicaltrials.gov registration. Judgement: high risk for reporting bias

Other bias

Low risk

Statistically significant differences between arms with respect to age, gender, and comorbidity at baseline. Judgement: Given the baseline imbalance, recruitment bias may potentially be present. We made a close‐call decision favouring low risk for other bias, as high risk for selection bias was already detected

Tattersall 2014

Methods

Parallel‐group randomised controlled trial (RCT)

Participants

Country and regions: Australia, Department of Medical Oncology, Royal Prince Alfred Hospital (RPAH) Camperdown, New South Wales

Recruitment: 22 months, no additional details reported

Inclusion criteria: newly detected incurable metastatic cancer (just diagnosed or relapsed with metastatic disease after previous adjuvant chemotherapy); life expectancy < 12 months (oncologist estimate of patient's likely survival time)

Exclusion criteria: previous contact with palliative care

Number of participants enrolled: N = 120 (60 intervention and 60 control) randomised

Participant characteristics: N = 107; mean age (intervention/control in years): 63/64; male gender (intervention/control in %): 53/43; married or living with partner (intervention/control in %): 67/68; education ≤ 10 years (intervention/control in %): 38/53; oncologist estimate of participant's likely survival time (intervention/control in %): 4‐12 weeks 2/0, 3‐6 months 15/10, 6‐12 months 55/50, > 12 months 18/33, not stated 10/7; intervention group with significantly more recent initial diagnosis and significantly more participants with likely survival time > 12 months

Diseases (intervention/control in %): lung cancer 20/18; gastrointestinal tract 33/40; breast 8/20; other gynaecological tumour 18/13; prostate 0/3; other tumour 20/5; differences between intervention and control not statistically significant

Deaths at end of study (intervention/control in N (%)): 39 (65)/31 (51.7); differences between intervention and control not statistically significant

Withdrawals/other drop‐outs (intervention/control in N (%)): 36 (60.0)/37 (61.7); differences between intervention and control not statistically significant

Interventions

Name: early contact with a palliative care nurse consultant with ongoing oncologist care vs oncologist care alone

Service base: outpatient palliative care

Intervention condition (n = 60): meeting with a palliative care nurse consultant member of the hospital palliative care team, who outlined available palliative care services including advice about symptom control, offered to arrange review by a palliative care physician, and provided contact details for the palliative care service; palliative care nurse offered to telephone the participant monthly to check on his or her well‐being, or, if the participant preferred, provided contact details for use by participant; standard oncological care given consistent with the oncologist’s recommendation
Control condition (n = 60): referral to the palliative care service when recommended by the oncologist

Outcomes

Primary endpoints: symptom severity (Rotterdam Symptom Checklist, RSC), quality of life (McGill Quality of Life Questionnaire, MQOL), degree of perceived support (Supportive
Care Needs – Short Form questionnaire, SCNS‐SF34)

Secondary endpoints: hospital medical records including end‐of‐life experiences, number of lines of chemotherapy, place of death

Assessment points: baseline/T0: after randomisation; T1: 1 month from enrolment; T2: 3 months from enrolment; T3: 6 months from enrolment; T4: 9 months from enrolment; T5: 12 months from enrolment

Notes

Funding source: National Health & Medical Research Council strategic palliative care research grant no. 219141

Declarations of interest among primary researchers: Apart from funding, no additional study author disclosure statements were made

Power considerations: At study completion, final enrolment was 120 owing to slower accrual than was projected in the initial power calculation (target sample size of 150, 80% power on a t test comparing treatment groups at a 2‐sided alpha of .05, effect size 0.50). Reduced sample size and power might have increased probability of type II error

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from main publication: "For allocation of the participants, a computer‐generated list of random numbers was used, and allocation was concealed using sequentially numbered, opaque sealed envelopes. No stratification was made for oncologist or cancer diagnosis"

Judgement: probably done

Allocation concealment (selection bias)

Low risk

Quote from main publication: "allocation was concealed using sequentially numbered, opaque sealed envelopes"

Judgement: probably done

Blinding of participants (performance bias)

High risk

Registration in the Australian New Zealand Clinical Trial Registry (ACTRN12611001137987) says it was an open‐label trial.

Judgement: not done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Original publication does not explicitly address blinding of outcome assessment

Incomplete outcome data (attrition bias)
All outcomes

High risk

Judgement: N = 21 completers in intervention group vs N = 29 completers in control group (Fisher's exact test with 1‐tailed P = 0.19). However, we assumed high risk of bias due to high loss of follow‐up across both groups

Selective reporting (reporting bias)

Unclear risk

Judgement: Outcomes from Australian New Zealand Clinical Trials Registry (ANZCTR) listed and reported in publications. However, trial registration was conducted after recruitment of participants. We made a close‐call decision favouring unclear risk against high risk of bias

Other bias

Unclear risk

Quote: "Most baseline characteristics were adequately balanced across the two study groups (Table 1), however there were differences between the groups in the time since initial cancer diagnosis (mean of 29 versus 34 months in the early referral and standard care groups respectively), and the oncologists’ estimate of likely survival (i.e. 11 versus 20 patients with estimates of > 12 months likely survival in the early referral and standard care groups respectively). Therefore, these variables were controlled for in subsequent analyses. There were no remarkable baseline differences on the patient reported outcome measures between the groups" Judgement: Given the baseline imbalance, recruitment bias may potentially be present. However, accounting for imbalance in statistical analysis did not change results. Thus, we made a close‐call decision favouring unclear risk against high risk of bias

Temel 2010

Methods

Parallel‐group randomised controlled trial (RCT)

Participants

Country and regions: USA, Massachusetts General Hospital, Boston, Massachusetts

Recruitment: 38 months; patients who presented to the outpatient thoracic oncology clinic were invited by their medical oncologists; all medical oncologists in the clinic agreed to approach, recruit, and obtain consent from their patients; physicians were encouraged, but were not required, to offer participation to all eligible patients; no additional screening or recruitment measures were used

Inclusion criteria: pathologically confirmed metastatic non‐small cell lung cancer; diagnosis within previous 8 weeks; Eastern Cooperative Oncology Group (ECOG) performance status of 0, 1, or 2; able to read and respond to questions in English

Exclusion criteria: patients already receiving care from the palliative care service

Number of participants enrolled: N = 151 (77 intervention and 74 control)

Participant characteristics: N = 151; mean age (intervention/control in years): 64.9/65.0; male gender (intervention/control in %): 51/45; married or living with partner (intervention/control in %): 61/62; Caucasian (intervention/control in %): 95/100; differences between intervention and control not statistically significant

Diseases (intervention/control in %): non‐small cell lung cancer 100/100; presence of brain metastases 26/31; receipt of initial chemotherapy as part of a clinical trial 27/21; never smoked or smoked ≤ 10 packs/y 22/24; illness perception of curable cancer 32/31; differences between intervention and control not statistically significant

Deaths at end of study (intervention/control in N (%)): 10 (13.0)/17 (23.0); differences not statistically significant (P = 0.14)

Withdrawals/other drop‐outs (intervention/control in N(%)): 7 (9.1)/10 (13.5); differences not statistically significant

Interventions

Name: early palliative care integrated with standard oncological care as compared with standard oncological care alone. All participants continued to receive routine oncological care throughout the study period

Service base: outpatient palliative care

Intervention condition (n = 77): "Patients who were assigned to early palliative care met with a member of the palliative care team, which consisted of board‐certified palliative care physicians and advanced‐practice nurses, within 3 weeks after enrolment and at least monthly thereafter in the outpatient setting until death. Additional visits with the palliative care service were scheduled at the discretion of the patient, oncologist, or palliative care provider. General guidelines for the palliative care visits in the ambulatory setting were adapted from the National Consensus Project for Quality Palliative Care and were included in the study protocol. Using a template in the electronic medical record, palliative care clinicians documented the care they provided according to these guidelines. Specific attention was paid to assessing physical and psychosocial symptoms, establishing goals of care, assisting with decision making regarding treatment, and coordinating care on the basis of the individual needs of the patient"

Control condition (n = 74): "Patients who were randomly assigned to standard care were not scheduled to meet with the palliative care service unless a meeting was requested by the patient, the family, or the oncologist; those who were referred to the service did not cross over to the palliative care group or follow the specified palliative care protocol"

Outcomes

Primary endpoints: quality of life (Trial Outcome Index, TOI, as sum of scores on the Lung Cancer Subscale and on physical and functional well‐being subscales of the Functional Assessment of Cancer Therapy‐Lung, FACT‐L)

Secondary endpoints: mood (Hospital Anxiety and Depression Scale, HADS; Patient Health Questionnaire 9, PHQ‐9), healthcare use and end‐of‐life care (anticancer therapy, medication prescriptions, referral to hospice, hospital admissions, emergency department visits, date and location of death), aggressive care, participants' resuscitation preferences

Assessment points: baseline/T0: before randomisation; T1: 3 months from enrolment

Notes

Funding source: American Society of Clinical Oncology Career Development Award and philanthropic gifts

Declarations of interest among primary researchers: Apart from funding, no additional study author disclosure statements were made

Power considerations: Primary outcome was change in score on the TOI from baseline to 12 weeks. Study authors estimated that with 120 participants, the study would have 80% power to detect a significant between‐group difference in the change in TOI score from baseline to 12 weeks, with a medium effect size of 0.5, SD.24. Protocol was amended in August 2008 to allow for enrolment of 30 additional participants to compensate for loss of any participants to follow‐up

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

According to a reply received from the principal investigator, computer‐generated random sequence generation with no stratification was applied.

Judgement: probably done

Allocation concealment (selection bias)

High risk

According to a reply received from the principal investigator, no allocation concealment.

Judgment: not done

Blinding of participants (performance bias)

High risk

Preregistration on clinicaltrials.gov (NCT01038271) says it was an open‐label trial.

Judgement: not done

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Original publication does not explicitly address blinding of outcome assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Judgement: N = 60 completers in intervention group vs N = 47 completers in control group (Fisher's exact test with 2‐tailed P value at 0.07). Trend toward higher attrition in the control group

Selective reporting (reporting bias)

Low risk

Judgement: All outcomes from clinicaltrials.gov registration listed and reported in publications

Other bias

Low risk

None detected

Zimmermann 2014

Methods

Study design: parallel‐group cluster‐randomised controlled trial (cRCT)

Participants

Country and regions: Canada, Princess Margaret Cancer Centre, University Health Network Toronto, Ontario

Recruitment: 51 months, daily screening of participating oncology clinics by research personnel

Inclusion criteria: aged 18 years or older; stage IV cancer (refractory to hormonal therapy as additional criterion for breast or prostate cancer, patients with stage III cancer and poor clinical prognosis were included at the discretion of the oncologist); estimated survival of 6‐24 months (assessed by main oncologist); ECOG performance status of 0, 1, or 2; completed baseline measures

Exclusion criteria: insufficient English literacy; inability to pass the cognitive screening test (Short Orientation‐Memory‐Concentration Test score < 20 or > 10 errors)

Number of participants enrolled: N = 461 (228 intervention and 233 control), 12 clinics allocated to intervention and control groups, respectively

Participant characteristics: N = 461; mean age (intervention/control in years): 61.2/60.2; male gender (intervention/control in %): 40.4/46.4; married or living with partner (intervention/control in %): 68.4/71.7; education < 9 years (intervention/control in %): 8.0/10.3; employed (intervention/control in %): 19.7/25.3; differences between intervention and control not statistically significant

Diseases (intervention/control in %): lung cancer 24.1/19.7; gastrointestinal tract 32.5/27.9; genitourinary 11.8/21.9; breast 18.0/13.3; other gynaecological tumour 13.6/17.2; control group with significantly larger number of participants with genitourinary cancers .02

Deaths at end of study (intervention/control in N (%)): 26 (11.4)/44 (18.9); differences statistically significant at P = 0.02

Withdrawals/other drop‐outs (intervention/control in N (%)): 52 (22.8)/53 (22.7); differences between intervention and control not statistically significant

Interventions

Name: early intervention in patients with advanced cancer by a palliative care team vs standard cancer care

Service base: outpatient clinics, hospital service, home care

Intervention condition (n = 228): multi‐disciplinary approach to care addressing physical, psychological, social, and spiritual needs Outpatient clinics: palliative care physician and nurse; routine visits once monthly and more often if necessary; routine structured symptom assessment in clinic during every visit by palliative care nurse and physician; routine psychosocial assessment in clinic and discussion of goals of care, of participant and family support needs, and of participant and family coping and psychological distress; discussion of advance care planning according to participant and family readiness; routine telephone follow‐up by palliative care nurse after each visit; more often as needed by palliative care nurse and physician; 24‐hour on‐call service explained during first visit, provided by specialised palliative care physicians Hospital service: direct access to palliative care unit for symptom management; primary care by trained palliative care nurses and physicians; formal 10‐day training for staff at opening of palliative care unit and continued education by palliative care unit advanced practice nurse; follow‐up by palliative care team when admitted to non‐palliative care unit service at University Health Network. Home care: community care access centre services explained and offered during first visit, reassessed at each visit; routine communication with family physician and community care access centre; home palliative care physician was explained during first visit and was offered with ECOG performance status ≥ 3 or when participant requested

Control condition (n = 233): approach to care mainly via addressing physical needs Outpatient clinics: oncologist and oncology nurses; visits ad hoc and mainly based on chemotherapy or radiation schedule; no structured symptom assessment; no routine psychological assessment; follow‐up as needed and conducted by oncology nurse, rare access to oncologist; access to 24‐hour on‐call service (oncology resident or clinical associate). Hospital service: no access to palliative care unit; admission to oncology ward or medical ward (via emergency department for urgent care); primary care by oncology nurses and clinical associates; no formal palliative care training; no follow‐up by palliative care team. Home care: community care access centre services ad hoc; generally no home care referral until referral to palliative care team; rarely an ad hoc communication with family physician and community care access centre; no home palliative care physician

Outcomes

Primary endpoints: participant‐reported quality of life (Functional Assessment of Chronic Illness Therapy‐Spiritual Well‐Being, FACIT‐Sp and Quality of Life at the End of Life, QUAL‐E)

Secondary endpoints: symptom impact (Edmonton Symptom Assessment System, ESAS), participant interaction with nurses and doctors (Cancer Rehabilitation Evaluation System Medical Interaction Subscale, CARES‐MIS), satisfaction with care (family satisfaction with advanced cancer care, FAMCARE‐P16)

Assessment points: baseline/T0: after randomisation; T1: 1 month from enrolment; T2: 2 months from enrolment; T3: 3 months from enrolment; T4: 4 months from enrolment

Notes

Funding source: Canadian Cancer Society, Ontario Ministry of Health and Long Term Care

Declarations of interest among primary researchers: Study authors declared no competing interests

Caregiver assessment: data from caregivers collected for an exploratory substudy, publication pending

Power considerations: Initial sample size estimation showed that 380 participants (190 per group) would provide 80% power at the 2‐sided 5% level of significance to detect a between‐group difference in FACIT‐Sp of 0.45 SD (medium effect size) by the primary endpoint of 3 months. Sample size was recalculated in 2008, on the basis of observed SD (from aggregated baseline data of 245 participants), intracluster correlation coefficient, attrition, and adherence. Revised sample size was 450 participants (225 per group)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote from main publication: "Randomisation was done by the statistical team at Western University (London, ON, Canada) using a computer‐generated sequence, was in a 1:1 ratio, and was stratified by clinic size and tumour site [...]"

Judgement: probably done

Allocation concealment (selection bias)

High risk

Quote from main publication: "There was also selection bias, which is common in cluster‐randomised studies because of randomisation of clusters before consent of individuals. A larger number of patients declined participation in the intervention group because of lack of symptoms"

Judgement: probably not done

Blinding of participants (performance bias)

Low risk

Quote from main publication: "Although complete masking of interventions was not possible, patients provided written informed consent to participate in their own study group, without being informed of the existence of another group. This form of masking is common in cluster randomised trials,

and avoids potential bias from patients in the control group requesting the intervention or otherwise altering their behaviour"

Judgement: probably done

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Quote from main publication: "Oncologists and investigators were aware of assignment"

Judgement: probably not done

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Judgement: N = 131 completers in intervention group vs N = 155 completers in control group (Fisher's exact test with 2‐tailed P value at 0.05). By conventional criteria, higher attrition in the intervention group of borderline significance. We made a close‐call decision favouring low risk against high risk of bias

Selective reporting (reporting bias)

Low risk

Judgement: Functional Assessment of Cancer Therapy‐General (FACT‐G) as primary outcome in clinicaltrials.gov registration, FACIT‐Sp (includes FACT‐G) reported as primary outcome in publications so far. Secondary outcomes Caregiver Quality of Life Index‐Cancer (CQOL‐C) and SF‐36 not reported in publications so far. However, all key outcomes have been reported

Other bias

Low risk

Tendency for higher outcome measure scores (for FACIT‐Sp at P = 0.03; for ESAS at P < 0.001; for FAMCARE‐P16 at P < 0.001) in intervention group at baseline. Larger number of participants with genitourinary cancers in the control group at baseline. No loss of clusters reported

Judgement: Given the baseline imbalance, recruitment bias may potentially be present. We made a close‐call decision favouring low risk for other bias, as high risk for selection bias was already detected

g/dL: grams per decilitre

µg/mL: microgram per millilitre

N = number of participants

packs/y: packs per year

PC: palliative care

SD: standard deviation

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Badr 2015

Psychosocial intervention focussing on dyadic coping. No genuine multi‐dimensional palliative care approach

Brumley 2007

No outcome measurement regarding symptom intensity or quality of life

Caruso 2014

Psychotherapy only. No genuine multi‐dimensional palliative care approach

Dyar 2012

Discussions of the benefits of hospice and advanced directives, led by a single nurse only. No genuine multi‐dimensional palliative care approach

Ferrell 2015

Non‐randomisation, instead quasi‐experimental trial with sequential enrolment of patients into control and intervention groups

Gade 2008

Study followed a conventional palliative care approach. No genuine early palliative care intent

Grudzen 2016

ED‐initiated palliative care consultation. No genuine early palliative care intent

Jensen 2014

Aerobic exercise, resistance or respiratory training only. No genuine multi‐dimensional palliative care approach

Jordhoy 2001

Portion of patient sample in the terminal phase. Study followed a conventional palliative care approach. No genuine early palliative care intent

Laing 1975

Randomised prospective trial comparing "no immediate treatment" with single‐ and multiple‐agent chemotherapy. However, study did not follow a proactive palliative care intent as characteristic for early palliative care

Lloyd‐Williams 2013

Psychosocial intervention applying narrative interviews. No genuine multi‐dimensional palliative care approach

Mok 2012

Portion of patient sample in the terminal phase. Study followed a conventional palliative care approach. No genuine early palliative care intent

NCT02311465

Investigators withdrew the study before enrolment

Pantilat 2010

Study followed a conventional palliative care approach. No genuine early palliative care intent

Rabow 2004

Patient sample had a life expectancy of 1 to 5 years and varying diseases. No genuine early palliative care intent

Rummans 2006

No explicit focus on physical domain/symptom control. No genuine early palliative care intent

Schofield 2013

Portion of patient sample in the terminal phase. Study followed a conventional palliative care approach. No genuine early palliative care intent

Stein 2005

Psychosocial intervention only. No genuine multi‐dimensional palliative care approach

Thoonsen 2011

Ongoing implementation study in which general practitioners were randomised. We do not consider this study to be a clinical trial on a patient population

Toseland 1995

Psychosocial intervention only. No genuine multi‐dimensional palliative care approach

Young 2013

Telephone follow‐up intervention for postoperative patients with colorectal cancer. Most patients with prognosis longer than 24 months. No genuine early palliative care intent

Characteristics of studies awaiting assessment [ordered by study ID]

Aljohani 2015

Methods

RCT

Participants

Patients with newly diagnosed NSCLC have a high symptom burden, poor quality of life, and a prognosis less than 1 year

Interventions

Early palliative care integrated with standard oncological care

Outcomes

Primary outcome measures: quality of life at 12 weeks assessed with the ESAS

Notes

Abstract with results published. We found no preregistration entry for this study

Groenvold 2017

Methods

RCT

Participants

Patients in contact with oncology departments who had cancer stage IV according to the ‘TNM’ (tumour, node, metastases) classification or cancer in the central nervous system grade 3 or 4, were at least 18 years of age, lived in the area of one of the participating specialised palliative care centres, and had not had contact with an SPC during the previous year received a screening questionnaire. If, according to their answers on the questionnaire, patients had a palliative need and 4 additional symptoms (see definition below), they were informed about the study and were invited to participate. Patients were excluded from the study if they could not understand Danish well enough to complete a questionnaire or were considered incapable of complying with the study protocol

Interventions

Experimental condition: specialised palliative care

Control condition: standard care

Outcomes

All randomised participants are assessed at baseline (the screening); after a 3‐week follow‐up period; and after an 8‐week follow‐up period. The primary outcome is estimated as the difference between intervention and control groups in the change from baseline to the weighted mean of the 3‐ and 8‐week follow‐up measured as area under the curve for the EORTC QLQ‐C30 scale score that constitutes the primary need. The primary need is defined as the palliative need having the highest intensity at baseline according to the EORTC QLQ‐C30. Secondary outcomes, estimated in the same way, are remaining symptoms and problems measured by the EORTC QLQ‐C30 (14 scales); anxiety and depression measured by the HADS; participants' evaluation of treatment and care provided by the healthcare system and measured by FAMCARE‐P16; survival; and economical consequences per week from the start of the study to minimum 3 months after the end of the intervention

Notes

Study completed. Protocol published

Kim 2016

Methods

Non‐randomised parallel assignment

Participants

Inclusion criteria:

  • Diagnosis of stage I‐IIIA resectable NSCLC ‐ undergoing lobectomy, pneumonectomy, segmentectomy, or wedge resection

  • Living within a 50‐mile radius of the City of Hope

  • No previous cancer within the past 5 years

Exclusion criteria:

  • Diagnosis of stage II‐III NSCLC that is not resectable based on clinical and individual characteristics (comorbidities, extent of disease, bulky mediastinal lymph nodes (N2), etc.)

  • Patients with NSCLC receiving radiofrequency ablation

Interventions

Experimental condition: group II (palliative care intervention); participants receive an individualized interdisciplinary palliative care intervention combining patient‐centred teaching principles and concepts that are learner‐entered (builds on strengths, interests, and needs of the learner), knowledge‐entered (teacher is proficient in the content being taught), assessment‐entered (learners are given an opportunity to test their understanding and receive feedback), and community‐entered (opportunities are available for continued learning and support); patients undergo 4 teaching sessions (based on patient‐entered teaching principles and concepts) that focus on physical, psychological, social, and spiritual well‐being, respectively, once a week in weeks 3‐6; patients then receive 4 follow‐up phone calls in weeks 9‐21 to clarify questions or review concerns from teaching sessions and to co‐ordinate follow‐up resources as needed

Control condition: group I (standard care); participants receive standard care; participants complete questionnaires at baseline and at 6, 12, 24, 36, and 52 weeks to evaluate quality of life (QOL), symptoms, psychological distress, and geriatric assessments

Outcomes

Primary outcome measures: overall quality of life and psychological distress at 6 months; symptom control at 6 months; geriatric assessment outcomes (OARS (Older Americans' Resources and Services) Instrumental Activities of Daily Living, MOS (Medical Outcomes Study) Activities of Daily Living, MOS Social Activities Limitation Scale, HADS scores, and Karnofsky performance scale); resource use (chart audits)

Notes

Study completed and results published.

Meyers 2011

Methods

Multi‐site RCT

Participants

Inclusion criteria:

  • Adults with relapsed, refractory, or recurrent solid tumours or lymphoma

  • Enrolled onto phase 1 or 2, or phase 3 trials that compared therapy for advanced cancer

Exclusion criteria:

  • Patients receiving concomitant chemotherapy and radiation

  • Patients on adjuvant phase III studies

  • Patients with hematopoietic malignancies

  • Patients with primary brain tumours

  • Patients not fluent in English

  • Patients < 18 years of age or lacking a willing caregiver

Interventions

Experimental condition: "intervention arm dyads received a copy of The Home Care Guide for Cancer. Each book chapter addresses a problem
known to affect patients with cancer including physical symptoms (pain or nausea), psychological symptoms (anxiety or depression), or issues related to resources or relationships, including communicating with one’s health care team or getting support or services from family, friends, and community organizations. Each chapter follows the same problem‐solving formula..Each educational session included the trained educator, the patient, and [the] designated caregiver. The first educational session was conducted up to 7 days prior to or on the day the patient started [the] investigational clinical trial. The first session focussed on becoming familiar with the guide and the COPE (Creativity, Optimism, Planning and Expert information) problem‐solving model, using COPE to address a patient or caregiver‐identified problem. The two additional conjoint instructional sessions were conducted within the first 30 days, reinforcing this learning by focusing on two additional patient or caregiver‐identified problems. Dyads could use one of the problems in the book’s chapters, or identify another problem and apply the model. In either event, the instructors facilitated this process of using the Guide and the COPE model, being careful not to solve the problem for them. Following each session, the educator documented the problem and recorded process notes"

Control condition: "usual care"

Outcomes

Primary outcome measures: City of Hope (COH) quality of life (QOL) instruments for patients or caregivers and the social problem solving inventory revised

Notes

Study completed. Results published. We contacted study authors for explicit information on palliative care intent. The reply to this study author request is pending

NCT00823732

Methods

Non‐randomised parallel assignment

Participants

Inclusion criteria:

  • Confirmed metastatic lung cancer (NSCLC, small cell lung cancer, and mesothelioma) or non‐colorectal gastrointestinal cancer (oesophageal, gastric, and hepatobiliary) not being treated with curative intent

  • Informed of metastatic disease within previous 8 weeks

  • No prior therapy for metastatic disease

  • Able to read questions in English or willing to complete questionnaires with the assistance of an interpreter

  • Relative or friend of participant who will likely accompany the participant to clinic visits

Exclusion criteria:

  • Significant psychiatric or other comorbid disease

Interventions

Experimental condition: phase 2 intervention GROUP II (palliative care intervention); participants receive an individualised interdisciplinary palliative care intervention comprising learner‐centred, knowledge‐centred, assessment‐centred, and community‐centred concepts; participants undergo 4 teaching sessions, focussed on physical, psychological, social, and spiritual well‐being, once weekly in weeks 3‐6; participants then receive 4 follow‐up phone calls in weeks 9, 13, 17, and 21

Control condition: no Intervention; phase I usual care GROUP I (usual care); participants receive standard care

Outcomes

Primary outcome measures: overall quality of life and psychological distress at 6 months; symptom control at 6 months; geriatric assessment outcomes (OARS (Older Americans' Resources and Services) Instrumental Activities of Daily Living, MOS (Medical Outcomes Study) Activities of Daily Living, MOS Social Activities Limitation Scale, Hospital Anxiety and Depression Scale scores, and Karnofsky performance scale); resource use (chart audits)

Notes

Study completed. Publication in preparation according to study authors

NCT01444157

Methods

RCT

Participants

Inclusion criteria:

At least 1 of the following

  • Cancer stage III or IV (according to hospital journal) and at least 1 treatment after relapse without satisfying effect on the disease

  • Patient is aware that further treatment is of palliative or life‐prolonging nature

And also all of the following inclusion criteria:

  • Patient has a family member who would like to participate (family member must be involved in patient care at least 2 times a week)

  • At least 18 years old (patient and family member)

  • Understand and speak Danish (patient and family member)

  • Live in the area of the municipalities of Copenhagen or Frederiksberg

  • Discharge from hospital to own home

  • Written informed consent (patient and family member)

Exclusion criteria:

  • Terminal phase of disease

  • Contact with specialised palliative care

  • Incapable of co‐operating with study protocol

  • Participant in another behavioural intervention study

Interventions

Experimental condition: palliative home care nursing group; in addition to standard home care nursing, families will receive 6 home visits from a research nurse with at least 1 year specialised palliative care experience; during the first 2‐hour visit, a family assessment is obtained that identifies family roles, resources, and coping strategies; the first home visit takes place no later than 1 week after randomisation; visits continue every third week up to 16 weeks, each visit with a duration of 1.5 hours; at every visit, the EORTC‐QLQ‐C30 patient‐administered questionnaire is used to identify the nature, frequency, and intensity of the patient's physical and psychosocial problems

Control condition: standard home care nursing group; patients continue to receive standard home care nursing; they can contact municipality services for visitation to home care nursing if they feel that additional home care is needed, or if they do not yet receive this service and feel they need home care nursing

Outcomes

Primary outcome measures: participant‐reported health‐related quality of life (EORTC QLQ‐C30 in relation to the global health status scale) at baseline, week 9, week 16, and week 24
Secondary outcome measures: participant‐reported symptoms and problems (EORTC QLQ‐C30 in relation to its functional scales and symptom scales/items) at baseline, week 9, week 16, and week 24; participant and family member symptoms of anxiety and depression (Hospital Anxiety and Depression Scale) at baseline, week 9, week 16, and week 24; family members' health‐related quality of life (SF‐36) at baseline, week 9, week 16, week 24, and 12 months; family satisfaction with healthcare services provided to the participant (FAMCARE) at baseline, week 9, week 16, week 24, and 12 months; acute readmission to hospital at weeks 16 and 24

Notes

Study completed. According to the principal investigator, publications are in preparation

NCT02133274

Methods

RCT

Participants

Inclusion criteria:

  • Age ≥18 years and <75 years

  • Adequate knowledge about the cancer diagnosis

  • Starting first‐line palliative antineoplastic treatment

  • Eastern Cooperative Oncology Group Performance Status ≤ 2

  • Life expectancy > 6 months and < 24 months (as per the medical oncologist)

  • Must have one of the following diagnoses:

    • metastatic or unresectable recurrent breast cancer

    • stage IIIC or IV recurrent platinum‐resistant ovarian cancer

    • metastatic or unresectable recurrent cervix cancer

    • metastatic or unresectable recurrent endometrial cancer

    • metastatic or unresectable recurrent head and neck cancer (after previous radiotherapy)

    • hormone‐refractory metastatic or unresectable recurrent prostate cancer

    • metastatic or unresectable recurrent genitourinary cancer

    • metastatic or unresectable recurrent non‐small cell lung cancer

    • extensive‐stage or recurrent small cell lung cancer

    • metastatic or unresectable recurrent gastrointestinal cancer

Exclusion criteria:

  • Currently undergoing any psychological treatment owing to a psychological disorder

  • Currently using antidepressants to treat depressive disorders and/or anxiety

  • Any cognitive deficit or attention problem that could interfere with ability to complete questionnaires or understand study aims (as per investigator)

  • Current or previously established diagnosis of any of the following psychological conditions: substance‐related disorders; schizophrenia and other psychotic disorders; mood disorders (depressive disorders, bipolar disorders); anxiety disorders; dissociative disorders; personality disorders; and/or history of a suicide attempt

  • Patients with single resected metastasis

  • Any comorbid condition, which, in the opinion of the investigator, could interfere with safety, compliance with the study, or interpretation of the results

  • Patients unable to go to the hospital for study visits, regardless of the reason

Interventions

Experimental condition 1: early palliative care; a first medical consult at the palliative care service will be scheduled after 2 to 3 weeks from study inclusion and every 3 to 4 weeks thereafter

Experimental condition 2: psychosocial plus early palliative care; 5 weekly sessions of a brief psychosocial intervention based on cognitive‐behavioural therapy plus early palliative care; regarding early palliative care, a first medical consult at the palliative care service will be scheduled after 2 to 3 weeks from study inclusion and every 3 to 4 weeks thereafter

Control condition: no Intervention; standard oncological care

Outcomes

Primary outcome measures: change from baseline in depression symptoms (HADS, PHQ‐9) at day 90; change from baseline in satisfaction with care on the FAMCARE‐patient scale at days 45, 90, 120, and 180; descriptive results about feasibility of the study
Secondary outcome measures: change from baseline in depressive symptoms on HADS‐D and PHQ‐9 at days 45, 120, and 180; change from baseline in anxiety symptoms on the HADS‐A at days 45, 90, 120, and 180; proportion of participants answering that their cancer is curable as measured with an adapted instrument to evaluate cancer understanding at 90, 120, and 180 days; change from baseline in cancer symptoms on the ESAS‐br at days 45, 90, 120, and 180; change from baseline in quality of life on the EORTC QLQ‐C15‐Pal at days 45, 90, 120, and 180

Notes

Last updated on ClinicalTrials.gov on 12 February 2017: "This study has been terminated. Planned interim analysis did not show the expected benefit of intervention A over B (effect size <0.2)."

NCT02207322

Methods

RCT

Participants

Inclusion criteria:

  • Patient eligibility criteria: adult patients (≥ 18 years) with haematological malignancy admitted to MGH for HSCT; ability to speak English or able to complete questionnaires with minimal assistance required from an interpreter or family member

  • Caregiver eligibility criteria: adult caregivers (≥ 18 years) of patients undergoing HSCT at MGH who agreed to participate in study; a relative or a friend, identified by the patient, who lives with the patient or has in‐person contact with him or her at least twice per week; ability to read questions in English or willing to complete questionnaires with the assistance of an interpreter

Exclusion criteria:

  • Patients with prior history of HSCT

  • Patients undergoing HSCT for a benign haematological condition (myelodysplastic syndrome is not considered a benign haematological condition and patients with myelodysplastic syndrome are eligible for the study)

  • Significant uncontrolled psychiatric disorder (psychotic disorder, bipolar disorder, major depression) or other comorbid disease (dementia, cognitive impairment), which the treating clinician believes prohibits informed consent or participation in the study

  • Patients enrolled in other supportive care intervention trials

Interventions

Experimental condition:

  • Standard transplant oncology care with early palliative care

  • Participant enrolment and caregiver enrolment (within 72 hours of participant enrolment)

  • Longitudinal data collection (participant and family caregivers)

Control condition:

Standard transplant oncology care with participant enrolment and caregiver enrolment (within 72 hours of participant enrolment) and longitudinal data collection (participants and family caregivers)

Outcomes

Primary outcome measures: change in FACT‐BMT score at 2 weeks, comparison of changes in quality of life (FACT‐BMT) scores from baseline to week 2 (day+5 for autologous, day+8 for myeloablative or reduced intensity allogeneic HSCT) between study arms by the 2‐sample t‐test

Notes

Study completed. Abstract with results published. Last updated on ClinicalTrials.gov on 28 January 2017: "This study is ongoing, but not recruiting participants."

Temel 2017

Methods

RCT

Participants

Inclusion criteria:

  • Confirmed metastatic lung cancer (NSCLC, small cell lung cancer, and mesothelioma) or non‐colorectal gastrointestinal cancer (oesophageal, gastric, and hepatobiliary) not being treated with curative intent

  • Informed of metastatic disease within previous 8 weeks

  • No prior therapy for metastatic disease

  • Able to read questions in English or willing to complete questionnaires with the assistance of an interpreter

  • Relative or friend of patient who will likely accompany the patient to clinic visits

Exclusion criteria:

  • Significant psychiatric or other comorbid disease

Interventions

Experimental condition: early palliative care; participants receive standard care with early palliative care

Control condition: no intervention; participants receive standard of care

Outcomes

Primary outcome measures: quality of life (FACT) at baseline and at 12 weeks
Secondary outcome measures: quality of life (FACT) with change from baseline over 24 weeks; mood (HADS) at baseline, and at 12 and 24 weeks; prognostic understanding at baseline, and at 12 and 24 weeks; family caregiver quality of life (SF‐36) at baseline, and at 12 and 24 weeks; family caregiver mood (HADS) at baseline, and at 12 and 24 weeks; family caregiver prognostic understanding at 12 and 24 weeks; resource utilisation at the end of life; chemotherapy utilisation at the end of life; hospice utilisation; healthcare costs; code status documentation; coping (Brief Cope) at baseline, and at 12 and 24 weeks; lung cancer‐specific quality of life (FACT‐Lung) at 12 and 24 weeks, GI cancer‐specific quality of life (FACT‐hepatobiliary and FACT‐espophageal) at 12 and 24 weeks
Other outcome measures: additional resource utilisation, hospital admissions, emergency room admissions, intensive care unit admissions, resuscitation attempts, survival

Notes

Study completed and results published.

Van Arsdale 2016

Methods

RCT

Participants

All patients defined as having high risk of gynaecological malignancies (< 30% 5‐year predicted survival)

Interventions

Referred to palliative care consultation within 8 weeks of tumour board registration for primary occurrence or recurrence

Outcomes

Primary outcome measure: increase in palliative consultation from historical 50% as reported at the tertiary care institution

Notes

Study completed. Abstracts with results published,

EORTC QLQ‐C30: European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire

EORTC QLQ‐C15‐Pal: European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire for Palliative Cancer Care Patients

ESAS: Edmonton Symptom Assessment System

FACT: Functional Assessment of Cancer Therapy

FACT‐BMT: Functional Assessment of Cancer Therapy ‐ Bone Marrow Transplant

FAMCARE: Family Satisfaction with Advanced Cancer Care Questionnaire

HADS: Hospital Anxiety and Depression Scale

HSCT: Haematopoietic stem cell transplantation

MGH: Massachusetts General Hospital (MGH)

NSCLC: non‐small cell lung cancer

PHQ‐9: Patient Health Questionnaire

RCT: randomised controlled trial

SF‐36: Short Form (36) Health Survey

Characteristics of ongoing studies [ordered by study ID]

ACTRN12610000724077

Trial name or title

Improving communication and quality of life (QOL) at the end of life: a randomised controlled trial of a multifocal communication intervention for patients with advanced incurable cancer, carers and doctors

Methods

Parallel‐group RCT

Participants

Patients are eligible if they have been given the diagnosis of any type of cancer, and their medical oncologist believes they have a life expectancy of between 2 and 12 months. Caregivers are eligible if they are identified as the primary, informal providers of care to a patient participating in the study. Patients and caregivers must read and speak English well enough to be interviewed and to complete questionnaires without the aid of an interpreter, must be over the age of 18 years, and must be capable of giving informed consent. Patients and caregivers will be excluded if they do not speak English or have significant psychological morbidity or cognitive impairment

Interventions

Guided by the self‐determination theory of health‐behaviour change, the communication support programme pairs a purpose‐designed Question Prompt List (an evidence‐based list of questions participants/caregivers can ask clinicians) with nurse‐led exploration of Question Prompt List content, communication challenges, participant values and concerns, and the value of early discussion of end‐of‐life issues. Oncologists are also cued to endorse participant and caregiver questions and use of the QPL. Behavioural and self‐report data will be collected from participants/caregivers approximately quarterly for up to 2.5 years, or until participant death, after which participant medical records will be examined. Analyses will examine the impact of the intervention on participants' and caregivers’ participation in medical consultations, their self‐efficacy in medical encounters, quality‐of‐life, end‐of‐life care receipt, and quality‐of‐death indicators

Outcomes

Patients: demographic details, communication self‐efficacy (Perceived Efficacy in Patient Physician Interactions Scale), quality of life (FACT‐G, and the McGill Quality of Life Scale), preferences for information and involvement in decisions about care as well as achievement of preferences for information and involvement in decisions (Degner Control Preference Scale, Cassileth Information Styles Questionnaire), hopes for treatment, preferences for future interventions, acceptance of disease (Peace, Equanimity and Acceptance in the Cancer Experience Scale), understanding of prognosis, doctor’s communication skills and manner

Caregiver: demographic details, communication self‐efficacy (adapted version of the Perceived Efficacy in Patient Physician Interactions Scale), quality of life (SF‐36), preferences for information and involvement in decisions about patient care (adapted Degner Control Preference Scale, Cassileth Information Styles Questionnaire), achievement of preferences for information and involvement in decisions about care, understanding of participants' hopes for treatment, understanding of participants' preferences for future interventions, understanding of participants' prognosis, Quality of Death and Dying Scale.

Starting date

April 2010

Contact information

Prof Phyllis Butow

Centre for Medical Psychology & Evidence‐based Decision‐making

School of Psychology

Brennan MacCallum Building (A18)

University of Sydney NSW 2006

Australia

E‐mail: [email protected]

Notes

Recruiting. Protocol published.

CTRI/2013/11/004128

Trial name or title

A study to assess the feasibility of introducing early palliative care in ambulatory patients with advanced lung cancer

Methods

Feasibility study

Participants

Inclusion criteria:

  • Patients with advanced lung cancer (stage IV)

  • ECOG Performance Status 0, 1, and 2

  • Patients who can adhere to follow‐up schedule at TATA Memorial Hospital

  • Age > 18 years

  • Written informed consent

Exclusion criteria:

  • ECOG > 2

  • Expected survival < 4 weeks

Interventions

Experimental condition: pain and symptom management and participant counselling; participants meet the palliative care team on the day of referral, then every month thereafter for 6 months

Outcomes

Primary outcome measures: More than 60% of referred patients have met the palliative care team; more than 50% of referred patients have completed the EORTC QLQ‐30 and EORTC QLQ ‐ LC13 (lung‐ cancer) and ESAS

Secondary outcome measures: symptom burden (ESAS), quality of life (EORTC QLQ 30 and EORTC QLQ ‐ LC13)

Starting date

September 2013

Contact information

Dr Jayita Deodhar

Associate Professor

Tata Memorial Hospital

Dr. E. Borges Road Parel Mumbai

Mumbai, Maharastra, 400012

India

E‐mail: [email protected]

Notes

Last updated on CTRI on 22 May 2017: "open to recruitment"

CTRI/2016/03/006693

Trial name or title

Effect of early integration of specialized palliative care into standard oncologic treatment on the quality of life of patients with advanced head and neck cancers: a randomized controlled trial

Methods

Randomised, parallel‐group trial

Participants

Inclusion criteria:

  • Patients with histologically confirmed squamous cell carcinoma of head and neck cancer in stage IV

  • ECOG 0, 1, and 2

  • Planned for treatment with palliative intent

  • Age > 18 years

  • Understands Hindi, Marathi, or English

  • Willing to participate in follow‐up

Exclusion criteria:

  • Patients with surgically resectable tumours

  • Planned for definitive radiotherapy

  • Uncontrolled comorbidities, such as uncontrolled diabetes mellitus, uncontrolled hypertension

  • Already receiving care from palliative care services

Target sample size is N = 180

Interventions

Experimental condition: early palliative care arm: Participant will receive specialist palliative care along with standard oncological treatment; participants in this arm will consult palliative care team along with parent oncology team; follow‐up visit will be provided as per need; chemotherapeutic drugs will be given according to disease status as per institutional protocol

Control condition: standard arm: Participants in this arm will receive standard oncological treatment; follow‐up visits will be decided by the oncologist as per treatment protocol; chemotherapeutic drugs will be given according to disease status as per institutional protocol

Outcomes

Primary outcome measures: change in quality of life, measured by FACIT‐H&N at 3 months after initial visit
Secondary outcome measures: changes in symptom burden assessed by Edmonton Symptom Assessment Scale (ESAS‐r) at 3 months after initial visit; overall survival at 3 months after initial visit

Starting date

March 2016

Contact information

M.A. Muckaden

Room 132, Department of Palliative Medicine, Ground Floor, Main Building, Tata Memorial Hospital, Parel (E), Mumbai 400012 Mumbai, MAHARASHTRA India

Notes

Last updated on CTRI on 22 May 2017: "not yet recruiting"

DRKS00006162

Trial name or title

Early palliative care – health services research and implementation of sustainable changes

Methods

Feasibility study

Participants

Early palliative care services provided in this study are aimed at patients with advanced metastatic cancer that is unresponsive to curative treatments (ICD 10 C 1–80 + ICD 10 C 78–79). In all participating comprehensive cancer centres, patients will be identified by the tumour boards at each centre. As soon as the diagnostic process has been concluded and treatment has started (i.e. within the first 8 weeks after diagnosis), patients will be referred to the PC physician

Target sample size is N = 2000

Interventions

In the main study phase, participants with metastatic cancer will routinely be offered a consultation with the palliative care physician within 8 weeks of diagnosis. This initial consultation has multiple objectives. First, this meeting serves to provide information regarding the value and accessibility of specialist palliative care. The palliative care physician will explain to participants that interdisciplinary cancer treatment ensures that all meaningful treatment options will continue to be available (“fight against cancer”), but that high priority will be placed on quality of life also. For quality of life needs, specialist palliative care services will be available to participants, alongside treatment from the primary cancer specialist

Outcomes

Early palliative care will be considered feasible if 75% of all eligible patients (i.e. adult patients with the diagnosis of an incurable, metastatic cancer [ICD 10 C 1–80 + ICD 10 C 78–79]) are referred to a palliative care physician at their centre at least once within 8 weeks of the initial diagnosis. Participants' quality of life and symptom burden will be assessed at the initial palliative care consultation on the POS, the EORTC QLQ‐C30, and the HADS. In both preliminary and main study phases, follow‐up assessment will be conducted at 12 and 24 weeks with these 3 instruments. Family/caregivers of the participant will be asked to assess the participant's situation by filling out the Quality of Dying and Death questionnaire.

Starting date

October 2014

Contact information

Dr Cornelia Meffert

Department of Palliative Care, Comprehensive Cancer Center

University Medical Center Freiburg

Robert‐Koch‐Str. 3

79106 Freiburg

Germany

E‐mail: cornelia.meffert@uniklinik‐freiburg.de

Notes

Protocol published. Last updated on DRKS on 22 May 2017: "recruiting"

ISRCTN13337289

Trial name or title

SPECIAL: Standard or palliative care in advanced lung cancer ‐ does early referral of patients with metastatic non‐small cell lung cancer to UK specialist palliative care services make a difference in their quality of life or survival?

Methods

Phase III randomised controlled trial with integral feasibility stage (non‐randomised)

Participants

Inclusion criteria:

  • Any adult (18 years or older) patient with newly diagnosed stage IV non‐small cell lung cancer, with histologically confirmed diagnosis

  • ECOG performance score 0‐3

Exclusion criteria:

  • ECOG performance score 4

  • Prognosis of 2 weeks

  • Participation in another local competing supportive or palliative care study

  • Dementia, delirium, or other lack of capacity or communication that renders the patient unable to participate in the study

  • Any other psychological disorder that, in the view of the investigator, renders the patient unable to participate

  • Unable to communicate in English or with the help of an interpreter

Interventions

Arm A: standard of care (i.e. standard referral to specialised palliative care, if participant is willing)
Arm B: sub‐randomisation
Arm B1: early specialised palliative care referral + standard of care
Arm B2: early specialised palliative care referral + standard of care + Sheffield Profile for Assessment and Referral for Care assessment

Outcomes

Primary outcome measures: Global Health Status Score at 3 months after study entry, quality‐adjusted survival time over 6 months

Secondary outcome measures: overall survival, anxiety/depression, pain, health economics, quality of life, memory and cognitive ability, Modified Glasgow Prognostic Score

Starting date

September 2015

Contact information

Sam H. Ahmedzai

c/o Trial Co‐ordinator Cancer Research (UK) Clinical Trials Unit

School of Cancer Sciences

University of Birmingham

Edgbaston

B15 2TT

Birmingham

United Kingdom

E‐mail: [email protected]

Notes

Last updated on ISRCTN on 28 November 2016: "recruiting completed"

ISRCTN18955704

Trial name or title

A multicentre non‐blinded randomised controlled trial to assess the impact of regular early specialist symptom control treatment on quality of life in malignant mesothelioma (RESPECT‐MESO)

Methods

Multi‐centre non‐blinded, randomised controlled, parallel‐group trial

Participants

Inclusion criteria:

  • Histological or cytological confirmation of malignant pleural mesothelioma

  • ECOG performance score of 0 to 1

  • Diagnosis of malignant pleural mesothelioma received within the last 6 weeks

  • Ability to provide written informed consent in English and comply with trial procedures

Exclusion criteria:

  • Other known malignancy within 5 years (excluding localised squamous cell carcinoma of the skin, cervical intraepithelial neoplasia, grade III, and low‐grade prostate cancer (Gleason score < 5, with no metastases))

  • Significant morbidity that the lead physician or multidisciplinary team feels will unduly confound or influence health‐related quality of life

  • Patients the multi‐disciplinary team judges require referral to specialist palliative care at the point of diagnosis

  • Concurrent, or within 3 months, participation in another clinical trial that may affect health‐related quality of life

  • Referral at the time of recruitment for cytoreductive, tumour de‐bulking, radical decortication or extrapleural pneumonectomy surgery for malignant pleural mesothelioma (video‐assisted thoracoscopic surgery or ‘mini’ thoracotomy for pleurodesis and diagnosis attempts are permissible)

  • Chemotherapy treatment for malignant pleural mesothelioma initiated before consent

  • Significant history of depression/anxiety/psychiatric illness requiring specialist hospital care within the last 12 months

Target sample size is N = 174

Interventions

Experimental condition: regular early SSCT: In the regular early SSCT group, participants will be seen within 3 weeks of randomisation by the specialised palliative care team (regardless of, and in addition to, all other treatments being offered). The initial meeting will consist of an approximately 1‐hour consultation with a member of the specialist palliative care team. This may be a Consultant or Specialist Palliative Care Clinical Nurse Specialist. Participants then will continue to be seen regularly on at least a 4‐weekly basis (regardless of other treatments, interventions, and symptoms) by a member of the specialist palliative care team, with consultations lasting approximately 30 minutes. These monthly reviews will continue until end of trial or participant death.

Control condition: "standard therapy"

Outcomes

Primary outcome measures: global quality of life at 12 weeks post randomisation

Secondary outcome measures:

  • Quality of life of participants after 24 weeks

  • Participant mood at 12 and 24 weeks

  • Primary caregiver quality of life and mood at 12 and 24 weeks, and at 24 weeks after participant death

  • Overall survival between study groups

  • Healthcare utilisation and healthcare costs

  • Cost‐effectiveness of regular early SSCT when compared with usual practice

Added 21/08/2014:

  • Subgroup analysis of health‐related quality of life at 12 and 24 weeks for participants based on neutrophil, lymphocyte ratio, and radiological staging at time of diagnosis

Starting date

January 2014

Contact information

Portsmouth Hospitals NHS Trust
Department of Respiratory Medicine
Queen Alexandra Hospital
Southwick Hill road
Cosham
Portsmouth
PO6 3LY
United Kingdom
+44 (0)23 9228 6000
E‐Mail: chief‐investigator‐ajc@respect‐meso.org

Notes

Protocol published. Last updated on ISRCTN on 17 October 2016: "completed"

NCT01589328

Trial name or title

Randomized controlled trials for the effect of early management on PAin and DEpression in patients with PancreatoBiliary cancer (EPADE‐PB)

Methods

RCT

Participants

Inclusion criteria:

  • Ages eligible for study: 18 years and older

  • Pathologically confirmed locally advanced or metastatic pancreatic cancer or biliary tract cancer

  • Within 8 weeks after diagnosis

  • Cancer‐related pain (BPI worst pain score > 3), depression (CES‐D > 16), or both

  • Karnofsky Performance Rating Scale ≥ 50%

Exclusion criteria:

  • Opioid intolerance

  • History of drug or alcohol abuse

  • Impaired sensory or cognitive function

  • Pregnant and lactating women

  • Women of child‐bearing potential not using a contraceptive method

  • Sexually active fertile men not using effective birth control during medication of study drug and up to 6 months after completion of study drug if their partners are women of child‐bearing potential

Target sample size is N = 288

Interventions

Experimental condition: early palliative care; interventions consisted of the following: nursing assessment of pain and depression mood; pain control‐based NCCN guideline; depression control by psychoeducation and/or consultation of psychiatrist specialist; participant education

Control condition: usual oncological care; participants randomly assigned to usual oncological care were not scheduled to meet with the palliative care service unless a meeting was requested by the participant, the family, or the oncologist; those who were referred to the service did not cross over to the early palliative care group or follow the specified palliative care protocol

Outcomes

Primary outcome measures: reduction in pain score (BPI) at 1 month and every 3 months up to 1 year; reduction in depression score (CES‐D) at 1 month and every 3 months up to 1 year
Secondary outcome measures: quality of life (EORTC QLQ‐C30 General Questionnaire, Korean version) at baseline, at 1 month, and every 3 months up to 1 year; overall survival

Starting date

April 2012

Contact information

WooJin Lee, MD

National Cancer Center

Goyang, 410‐769, Gyeonggi‐do

Republic of Korea

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 3 October 2016: "This study is currently recruiting participants."

NCT01828775

Trial name or title

Integration of palliative care for cancer patients on phase I trials

Methods

RCT

Participants

Inclusion criteria:

  • Patients diagnosed with solid tumours who are eligible for participation in phase I clinical trials of investigational cancer therapies

  • Patients who have signed an informed consent for participation in phase I clinical trials

  • Able to read or understand English ‐ this is included because the intervention and study materials (including outcome measures) are provided only in English

  • Ability to read and/or understand study protocol requirements and to provide written informed consent

Exclusion criteria:

  • Patients with diagnosis of haematological (as a population distinct from solid tumours and different studies) or brain cancers (due to cognitive ability)

Interventions

Experimental condition: early PCI; participants receive part I of the PCI comprising quantitative surveys, comprehensive palliative care assessment by research nurses, and goals of care discussions beginning before administration of the first dose of phase I treatment; participants then receive part II of the PCI comprising recommendations from the interdisciplinary team, participant educational sessions, and supportive care referrals following the first dose of phase I treatment and completed within 1 month of the first treatment

Control condition: delayed PCI; participants receive usual care until 12 weeks post treatment initiation; participants then receive both part I and part II of the PCI

Outcomes

Primary outcome measures: change in overall quality of life scores (FACT‐G and FACIT‐Sp) at 12 weeks; change in psychological distress (NCCN Distress Thermometer) at 12 weeks; satisfaction with communication (FAMCARE) at 12 weeks; participants' symptom intensity and symptom interference with daily activities (Psychological Patient‐Reported Outcomes Version of the Common Terminology Criteria for Adverse Events) at 4 and 12 weeks; total numbers of supportive care referrals (social work, dietician, chaplaincy, psychologist/psychiatrist) at 12 weeks; total numbers of unscheduled outpatient encounters and inpatient admissions at 12 weeks; total number of hospice referrals at 12 weeks; retention in the phase I trial at 12 weeks; participant satisfaction with the PCI at 12 weeks

Starting date

September 2014

Contact information

Betty Ferrell, PhD, MA, FAAN, FPCN

City of Hope ‐ Main Campus (Duarte)
1500 East Duarte Road
Duarte, CA 91010

United States

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 31 May 2017: "This study is currently recruiting participants."

NCT01865396

Trial name or title

Effect of early palliative care on quality of life of patients with advanced cancer: a randomised controlled trial

Methods

RCT

Participants

Inclusion criteria:

Patients with life‐limiting cancer (prognosis of approximately 1 year) are eligible if:

  • they are within 12 weeks of referral from another hospital after receiving first‐line treatment or within 8 to 12 weeks of a new diagnosis (histologically and cytologically confirmed): metastatic and advanced pancreatic, stomach, oesophageal, and biliary tract adenocarcinoma; metastatic or advanced NSCLC (stage IIIB or IV) or metastatic SCLC, malignant pleural mesothelioma, metastatic or advanced head and neck cancer (stage III or IV)

  • they are within 12 weeks of progression after receiving treatment and have a prognosis of approximately 1 year: metastatic and locally advanced colorectal cancer, with progression after second‐line treatment; metastatic or advanced prostate carcinoma after second‐line treatment, advanced breast cancer with visceral and/or brain metastasis, with progression on second‐ or third‐line treatment; metastatic melanoma, metastatic or advanced kidney cancer, metastatic or advanced bladder cancer after first‐line treatment

  • an ECOG performance status of 0, 1, or 2 and ability to read and respond to questions in Dutch

Exclusion criteria:

  • Patients < 18 years old

  • Patients with impaired cognition

  • Patients who met the palliative support team more than once or had a consultation within 6 months of inclusion

Target sample size is N = 186

Interventions

Experimental condition: early palliative care; interventional palliative care after diagnosis and once a month

Control condition: standard care; participants will receive standard oncological care

Outcomes

Primary outcome measures: quality of life of the participant and his or her family caregiver at baseline, at 12 weeks, and 6‐weekly after 12 weeks (EORTC‐QLQ C30, McGill QOL, SF‐36)
Secondary outcome measures: influence of palliative care on mood and illness understanding of participants and family caregivers at baseline, at 12 weeks, and 6‐weekly after 12 weeks (HADS, PHQ‐9, illness understanding), influence of palliative care on decisions of physicians with regards to end‐of‐life care (questionnaire for decisions with regards to end‐of‐life decision making for physicians)

Starting date

April 2013

Contact information

Gaëlle Vanbutsele, MSc, Clinical Psychologist Doctoral Researcher

End‐of‐Life Care Research Group

Vrije Universiteit Brussel & Ghent University

UZ Gent

De Pintelaan 185, 9000 Gent

Belgium

E‐mail: [email protected]

Notes

Abstract published. Last updated on ClinicalTrials.gov on 1 July 2016: "This study is ongoing, but not recruiting participants."

NCT01885884

Trial name or title

A pilot trial of an embedded collaborative model of supportive care for pancreatic cancer

Methods

RCT

Participants

Inclusion criteria:

  • Patients: adults (≥ 18 years old), pathologically confirmed locally advanced or metastatic pancreatic adenocarcinoma diagnosed within the past 8 weeks, ECOG Performance Status of 0 (asymptomatic), 1 (symptomatic but fully ambulatory), or 2 (symptomatic and in bed < 50% of the day), planning to receive continued care from an oncologist at the Hillman Cancer Center, accompanied by a caregiver (family member or friend) at the first visit

  • Caregivers: adults (≥ 18 years old), family member or friend of an eligible patient

Exclusion criteria:

  • Patients: unable to read and respond to questions in English, not planning to receive continued care from an oncologist at the Hillman Cancer Center Pancreatic, neuroendocrine cancer

  • Caregivers: unable to read and respond to questions in English

Target sample size is N = 60

Interventions

Experimental condition: supportive care intervention; monthly (minimum) participant and caregiver visits with a supportive care physician, embedded within their standard oncological care (through collaboration with oncology providers)

Control condition: usual care; participants will receive standard oncology care from their oncology providers

Outcomes

Primary outcome measures: trial feasibility; acceptability of intervention participation at 3 months (+/‐ 3 weeks); perceived effectiveness at 3 months (+/‐ 3 weeks)

Secondary outcome measures: change in participant quality of life (FACT‐Hep) at 3 months (+/‐ 3 weeks); participant healthcare utilisation (numbers and types of chemotherapy regimens, frequency and timing of chemotherapy regimens, number and length (days) of hospital admissions, number and length (days) of intensive care unit admissions, number of emergency department visits, frequency and timing (days before death) of hospice use, place of death)

Starting date

July 2013

Contact information

Yael Schenker, MD, MAS

Assistant Professor of Medicine

University of Pittsburgh Cancer Institute (UPCI)

Hillman Cancer Center

Pittsburgh, Pennsylvania, 15232

United States

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 10 May 2016: "This study has been completed."

NCT01983956

Trial name or title

A structured early palliative care intervention for patients with advanced cancer ‐ a randomized controlled trial with a nested qualitative study (SENS trial)

Methods

RCT

Participants

Inclusion criteria:

  • Diagnosed within the last 16 weeks

  • Metastatic or locally advanced, not amenable to curative treatment, NSCLC, or

  • Metastatic or locally advanced, not amenable to curative treatment, colorectal cancer, or

  • Metastatic or locally advanced, not amenable to curative treatment, prostate cancer, or

  • Metastatic or locally advanced, not amenable to curative treatment, breast cancer with visceral and/or brain metastasis, or

  • Metastatic or locally advanced, not amenable to curative treatment, bladder/urothelium cancer, or

  • Metastatic or locally advanced, not amenable to curative treatment, pancreatic cancer

  • Diagnosis histologically confirmed

  • ECOG Performance Status of 0, 1, or 2

  • At least 18 years of age at the time of enrolment

  • Signed informed consent with understanding of study procedures and the investigational nature of the study

Exclusion criteria:

  • Presence of delirium or dementia or other reason for lack of ability to give informed consent

  • Inability to communicate adequately in German

  • Lack of patient accountability; inability to appreciate the nature, meaning, and consequences of the study and to formulate his or her own wishes correspondingly

  • Patients already receiving care from an inpatient palliative care service

Target sample size is N = 150

Interventions

Experimental condition: structured approach intervention with the SENS model based on the bio‐psycho‐social‐spiritual model of care and WHO definitions of palliative care, as well as NCCN Practice Guidelines for Palliative Care. It supports assessment of areas and complexity of concerns from the participant perspective, determines the priority and structures the support needed. Intervention is provided by palliative care physicians and nurses collaboratively. It is utilised as baseline assessment and afterwards is integrated into each routine oncology care outpatient and inpatient visit. Depending on the goals, it may be applied between routine visits. In addition, participants will receive usual oncology care throughout the study period

Control condition: Participants in the usual care group will receive routine oncology care throughout the study. This incorporates a routine assessment according to the standard Swiss Group for Clinical Cancer Research (SAKK) protocol, which assesses overall symptoms. Participants are not seen by nurses during a routine visit to the outpatient clinic unless they need a blood withdrawal or any intravenous or subcutaneous treatment. Only nursing staff in the palliative care unit is familiar with using the SENS‐assessment instrument. Participants assigned to usual care may meet with the palliative care service on request according to established practice

Outcomes

Primary outcome measures: distress over 6 months (NCCN Distress Thermometer)
Secondary outcome measures: quality of life (FACT‐G) at 6 months; POS at 6 months; overall survival; location of death; healthcare utilisation (questionnaire of Stanford Patient Education Research Centre)

Starting date

December 2013

Contact information

Steffen Eychmueller, MD

University Center for Palliative Care

Bern University Hospital

SWAN Haus
Freiburgstrasse 28

CH‐3010 Bern

Switzerland

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 27 September 2016: "This study is currently recruiting participants."

NCT02308865

Trial name or title

Impact of early palliative care on quality of life and survival of patients with non‐small‐cell metastatic lung cancer in Northern France

Methods

RCT

Participants

Inclusion criteria:

  • Diagnosis of non‐small cell lung cancer, proven histologically, metastatic proven imaging (MRI, CT scanner, PET scan), stage IV (any T, any N, M1), diagnosed in the 8 weeks preceding inclusion, supported outpatient

  • Age > 18 years

  • PS ≤ 2

  • Patient able to understand the nature, purpose, and methods of the study

  • Signed Informed consent

Exclusion criteria:

  • Age < 18 years

  • Patient already supported by palliative care

  • Patient with an activating EGFR mutation or EML4‐ALK rearrangement

  • Patient under trusteeship/guardianship

Target sample size is N = 144

Interventions

Experimental condition: multidisciplinary palliative care monthly consultations with a doctor, a nurse, a psychologist, and possibility a physical therapist and a chaplain, in addition to standard onco‐pneumological care

Control condition: participant supported by the oncological respiratory service for treatment of disease by chemotherapy and for treatment of complications

Outcomes

Primary outcome measures: quality of life (Trial Outcome Index) at 12 weeks
Secondary outcome measures: survival; events (presence of any of the following: chemotherapy, use of resuscitation, or no treatment, limiting decision 14 days before death); quality of life (FACT‐L, PHQ‐9, and HADS questionnaires) at 12 and 21 weeks

Starting date

October 2014

Contact information

Licia Touzet, MD

University Hospital Lille

59000 Lille

France

E‐mail: licia.touzet@chru‐lille.fr

Notes

Last updated on ClinicalTrials.gov on 18 May 2017: "This study is currently recruiting participants."

NCT02332317

Trial name or title

A randomized, controlled phase III study of integrated, specialized palliative rehabilitation for patients with newly diagnosed non‐resectable cancer

Methods

RCT

Participants

Inclusion criteria:

Participants must:

  • receive a diagnosis of non‐resectable cancer less than 8 weeks before inclusion

  • be fit to receive standard oncology treatment and to accept treatment

  • read and understand Danish

  • sign informed consent

Exclusion criteria:

  • Contact with a specialised palliative unit within the last year before inclusion

  • Inability to co‐operate during the study

  • Missing informed consent

Target sample size is N = 300

Interventions

Experimental condition: 150 participants will receive standard oncology treatment alongside a 12‐week specialised palliative rehabilitation programme

Control condition: 150 participants will receive standard oncology treatment

Outcomes

Primary outcome measures: effect of the intervention on "The Primary Problem" chosen by the participant (EORTC‐QLQ‐C30 that correlates with "the primary problem" of the participant) at 6 and 12 weeks
Secondary outcome measures: EORTC‐QLQ‐C30 at 6 and 12 weeks; worries and symptoms of anxiety and depression (HADS) at 6 and 12 weeks; all‐cause mortality; economic consequences

Starting date

November 2014

Contact information

Lars Henrik Jensen, MD, PhD

Department of Oncology

VejleHospital

DK‐7100 Vejle

Denmark

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 31 May 2017: "This study is currently recruiting participants."

NCT02335619

Trial name or title

Early integrated supportive care study for gastrointestinal cancer patients

Methods

RCT

Participants

Inclusion criteria:

  • Diagnosis of gastrointestinal cancer

  • Appointments in gastronintestinal clinic during study days

  • Ability to complete a symptom assessment form alone or with the help of a family member or interpreter

Exclusion criteria:

  • Already receiving care from the Pain and Symptom Management/Palliative care team

Target sample size is N = 152

Interventions

Experimental condition: early palliative care; during first oncology appointment, participants in the intervention arm will self‐report to the study team any symptoms related to their cancer or treatment; scores at or above a defined benchmark will be seen by Pain and Symptom Management/Palliative Care team members during or immediately after their oncology appointment; participants will be asked to self‐report symptoms once a month following recruitment for 4 months

Control condition: standard care; during first oncology appointment, participants in the control arm will self‐report to the study team any symptoms related to their cancer or treatment; self‐reports will be collected but will not be shared with the Pain and Symptom Management/Palliative Care Team, and participants will continue with their oncology appointment as per standard procedure; participants will be asked to self‐report symptoms once a month following recruitment for 4 months

Outcomes

Primary outcome measure: total symptom distress score at 4 months after recruitment (modified ESAS)
Secondary outcome measures: use of health services at 4 months after recruitment (number of hospital admissions for non‐treatment reasons; number of emergency room visits; number of referrals to the Pain and Symptom Management/Palliative Care Team; number of Pain and Symptom Management/Palliative Care follow‐up visits per participant); aggressiveness of cancer treatment; details of death

Starting date

February 2015

Contact information

Pippa Hawley, MD

Head Palliative Care Physician

British Columbia Cancer Agency

Vancouver, British Columbia, V5Z 4E6

Canada

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 14 April 2015: "The recruitment status of this study is unknown. The completion date has passed and the status has not been verified in more than two years."

NCT02349412

Trial name or title

Methods

Participants

Interventions

The study intervention consists of early integration of palliative care services into standard oncology care provided in an outpatient setting for participants with advanced lung and non‐colorectal gastrointestinal malignancies who are not being treated with curative intent. Palliative care services provided to participants randomised to the intervention will be provided by board‐certified physicians and/or advanced practice nurses and will focus on the following areas: developing and maintaining the therapeutic relationship with participants and family caregivers; assessing and treating participant symptoms; providing support and reinforcement for coping with advanced cancer among participants and family caregivers; assessing and enhancing prognostic awareness and illness understanding in participants and family caregivers; assisting with treatment decision making; and (6) assisting with end‐of‐life care planning

Outcomes

Primary outcome measure: change in FACT‐G scores from baseline to 12 weeks
Secondary outcome measures: change in quality of life on FACT‐G over time, depressive symptoms as per HADS, rate of anxiety symptoms as per HADS at 12 weeks and over time, change in illness understanding over time, change in quality of life on the SF‐36 over time, rate of referral, enrolment and length of stay in hospice, location of death, number of hospital and intensive care unit admissions and days, chemotherapy and radiation administration, overall survival, concordance between participant and family caregiver report of prognosis/curability

Starting date

April 2015

Contact information

Jennifer S. Temel, MD

Massachusetts General Hospital, 55 Fruit St.
Yawkey 7B, Boston, MA 02114

United States

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 19 April 2017: "This study is ongoing, but not recruiting participants."

NCT02547142

Trial name or title

Evaluation of the implementation of an early integrated palliative care program in the esophageal cancer population

Methods

RCT

Participants

Inclusion criteria:

  • Patients newly diagnosed or referred to the Esophageal Diagnostic Assessment Program with suspicious findings found to be oesophageal cancer

  • Patients who present with metastatic disease, defined as N3 lymph node involvement or distant metastatic deposits as confirmed on PET scan

  • Patients must have been notified by a member of their healthcare team of their prognosis and palliative categorisation, as noted in the patient chart, within 8 weeks of diagnosis

  • Patients may undergo oesophagectomy, stenting, brachytherapy, or palliative intent chemotherapy or radiotherapy as clinically indicated

Exclusion criteria:

  • Individuals unable to complete questionnaires with assistance

  • Patients presently undergoing neoadjuvant chemotherapy or radiotherapy for malignancy

  • Patients with recurrent oesophageal cancer

  • Patients who are referred back to the Esophageal Diagnostic Assessment Program for restaging after completing neoadjuvant therapy

Target sample size is N = 700

Interventions

Early palliative care (not further specified)

Outcomes

Primary outcome measure: quality of life
Secondary outcome measures: oesophageal cancer‐specific symptom score FACT‐E, PHQ‐9, HADS, participant survival post metastatic oesophageal cancer diagnosis

Starting date

October 2015

Contact information

Christian J Finley, MD, MPH, FRCSC

McMaster University

50 Charlton Avenue East, T‐2105
Hamilton, Ontario L8N 4A6

Canada

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 15 March 2016: "This study is currently recruiting participants."

NCT02631811

Trial name or title

Early palliative care in patients with acute leukaemia (Pablo Hemato)

Methods

RCT

Participants

Inclusion criteria:

  • > 18 years old

  • Acute lymphoblastic or myeloblastic leukaemia at first relapse and diagnosed within 8 weeks before inclusion

  • Patients for whom a curative strategy (transplant) is not considered

  • Patients older than 75 years at diagnosis

  • Informed signed consent

Exclusion criteria:

  • Inability to complete the questionnaire

  • Psychiatric disorders other than depression

  • Persons under guardianship

Target sample size is N = 40

Interventions

Experimental condition: Participants will be seen by palliative care team at least once a month until the 12th week, more often if needed. Symptoms and suffering will be assessed by a multi‐disciplinary palliative specialist team of physician, nurse, and psychologist. Physical, psychological, social, and existential suffering will be addressed

Control condition: "usual medical follow‐up"

Outcomes

Primary outcome measure:

  • Measure of quality of life [time frame: 12 weeks] [designated as safety issue: no] ‐ quality of life measured by FACT‐Leu questionnaire. Score for quality of life will be compared between groups

Secondary outcome measures:

  • Measure of symptom intensity [time frame: 12 weeks] [designated as safety issue: no]

  • Symptom intensity measured by ESAS questionnaire. Score for symptom intensity will be compared between groups

  • Measure of depression [time frame: 12 weeks] [designated as safety issue: no] ‐ score of depression measured by HADS questionnaire. Score of depression will be compared between groups

  • Measure of anxiety [time frame: 12 weeks] [designated as safety issue: no] ‐ score of anxiety measured by HADS questionnaire. Score of anxiety will be compared between groups

  • Measure of quality of the end of life [time frame: up to 9 months] [designated as safety issue: no] ‐ within the last month of life, several parameters will be studied to evaluate the quality of the end of life, such as number of admissions in the emergency unit

  • Overall survival [time frame: 9 months] [designated as safety issue: yes]

Starting date

November 2015

Contact information

Marilène FILBET, PU‐PH

Centre Hospitalier Lyon Sud

Pierre Bénite, France, 69495

E‐Mail: marilene.filbet@chu‐lyon.fr

Notes

Last updated on ClinicalTrials.gov on 11 December 2015: "This study is currently recruiting participants."

NCT02712229

Trial name or title

A primary palliative care intervention for patients with advanced cancer (CONNECT)

Methods

Cluster‐randomised controlled trial (cRCT)

Participants

Inclusion criteria:

Participants will be patients with advanced cancer receiving care at a participating clinic; their caregivers; their oncology staff nurses, oncologists, and practice managers. They will be

adults (≥ 21 years old); with metastatic solid tumours; the oncologist "would not be surprised if the patient died in the next year"; Eastern Cooperative Oncology Group performance status (ECOG PS) ≤ 2; planning to receive ongoing care from a participating oncologist and willing to be seen at least monthly

Exclusion criteria:

Inability to read and respond to questions in English; cognitive impairment or inability to consent to treatment, as determined by the patient's oncologist; inability to complete baseline interview; ECOG PS of 3 (capable of limited self‐care; confined to bed or chair > 50% of waking hours) or 4 (cannot carry on any self‐care; totally confined to bed or chair); haematological malignancy

Target sample size is N = 1486

Interventions

Experimental condition: CONNECT

CONNECT is a primary palliative care ‐ care management intervention led by existing oncology nurses. CONNECT is deployed through a series of nurse‐led encounters occurring before or after regularly scheduled oncology clinic visits. Based on best practices in palliative oncology care, the first visit focusses on establishing rapport, addressing symptom needs, and choosing a surrogate decision maker. Subsequent visits include additional focus on treatment preferences and future goals. CONNECT visits are guided by participant‐reported outcomes. During every CONNECT encounter, the nurse will work with participants and caregivers to complete and update individualized shared care plans. After every CONNECT visit, the nurse will discuss participants' symptoms, preferences, and goals with their oncologists via a mandatory check‐in session and will conduct a follow‐up call with the participant and/or caregiver.

Control condition: Usual care control. At clinics randomised to usual care, enrolled participants and caregivers will continue to receive supportive oncology care according to usual practice.

Outcomes

Primary outcome measure:

  • Quality of life ‐ participant [time frame: change from baseline to 3 months] [designated as safety issue: no] ‐ investigators will compare change in 3‐month FACIT‐Pal scores between enrolled participants at intervention clinics and enrolled participants at usual care clinics

Secondary outcome measures:

  • Symptom burden ‐ participant [time frame: change from baseline to 3 months] [designated as safety issue: no] ‐ investigators will compare change in 3‐month ESAS scores between enrolled participants at intervention clinics and enrolled participants at usual care clinics

  • Depression and anxiety symptoms ‐ participant [time frame: change from baseline to 3 months] [designated as safety issue: no] ‐ investigators will compare change in 3‐month HADS scores between enrolled participants at intervention clinics and enrolled participants at usual care clinics

  • Depression and anxiety symptoms ‐ caregiver [time frame: change from baseline to 3 months] [designated as safety issue: no] ‐ investigators will compare change in 3‐month HADS scores between enrolled caregivers at intervention clinics and enrolled caregivers at usual care clinics

  • Caregiver burden ‐ caregiver [time frame: change from baseline to 3 months] [designated as safety issue: no ] ‐ investigators will compare change in 3‐month Zarit Burden Interview‐Short scores between enrolled caregivers at intervention clinics and enrolled caregivers at usual care clinics

  • Healthcare utilisation [time frame: 1 year] [designated as safety issue: no ] ‐ to inform future dissemination efforts and aid in understanding of optimal financing models, investigators will calculate implementation costs of the intervention and will determine the effects of CONNECT on healthcare utilisation, including hospitalisations, chemotherapy use, and hospice use

  • Survival ‐ participants [time frame: 1 year] [designated as safety issue: no ] ‐ investigators will calculate survival time from date of enrolment using the Kaplan‐Meier method. We will use frailty models to assess for any effect of CONNECT on survival, while controlling for effects of clustering.

Starting date

April 2016

Contact information

Yael Schenker, MD, MAS

Assistant Professor of Medicine

University of Pittsburgh Cancer Institute (UPCI)

Hillman Cancer Center

Pittsburgh, Pennsylvania, 15232

United States

E‐mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 7 December 2016: "This study is currently recruiting participants."

NCT02730858

Trial name or title

Palliative and oncology care model In breast cancer

Methods

RCT

Participants

Inclusion criteria:

  • Metastatic breast cancer

  • Diagnosis of any of the following within the prior 4 weeks: leptomeningeal disease; progressive brain metastases after initial radiation therapy; triple negative breast cancer with progressive disease on first‐line chemotherapy; or currently admitted to the hospital or admitted to the hospital within the prior 4 weeks

  • Receiving cancer care at Massachusetts General Hospital Cancer Center

  • Ability to read and write in English

  • ECOG status between 0 and 2

Exclusion criteria:

  • Already receiving palliative care in the outpatient setting

  • Active, untreated, unstable, serious mental illness (e.g. active, untreated psychotic, bipolar, or substance‐dependence disorder) interfering with ability to participate

  • Cognitive impairment (e.g. delirium, dementia) interfering with ability to participate

  • Requires urgent palliative or hospice care

Target sample size is N = 120

Interventions

Experimental condition: Participants randomised to the intervention will receive collaborative care from palliative care and oncology for the remainder of their illness. The initial 5 visits with palliative care will be conducted in accordance with study‐specific clinical practice guidelines and will occur at least monthly

Control condition: Participants randomised to oncology care alone will continue to receive routine care identical to what they would have received if they had not participated in the trial. Participants or their oncologists can request palliative care consultation at any point in time

Outcomes

Primary outcome measure:

  • End‐of‐life care preference documentation [time frame: 1 year] [designated as safety issue: no] ‐ compare differences in rate of documentation of end‐of‐life care preferences (Yes documented vs No)

Secondary outcome measures:

  • Participant‐reported end‐of‐life care conversation [time frame: 6 months] [designated as safety issue: no] ‐ examine participant report of end‐of‐life care preferences with the clinician using the following item: "Have you and your doctors discussed any particular wishes you have about the care you want to receive if you were dying?" Although participants complete this measure repeatedly during the course of the study, investigators will use the final assessment before death or at 6 months' follow‐up (whichever comes first) for this analysis

  • Participant‐reported quality of life (FACT‐Breast) [time frame: weeks 6, 12, 18, and 24] [designated as safety issue: no] ‐ compare participant‐reported quality of life between study arms at weeks 6, 12, 18, and 24

  • Participant‐reported depression symptoms (HADS) [time frame: weeks 6, 12, 18, and 24] [designated as safety issue: no] ‐ compare participant‐reported depression symptoms between study arms at weeks 6, 12, 18, and 24

  • Participant‐reported anxiety symptoms (HADS) [time frame: weeks 6, 12, 18, and 24] [designated as safety issue: no ‐ compare participant‐reported anxiety symptoms between study arms at weeks 6, 12, 18, and 24

  • Chemotherapy at end of life [time frame: 14, 7 and 3 days before death] [designated as safety issue: no] ‐ examine differences in rates of chemotherapy administration during the last 3, 7, and 14 days of life between study arms

  • Rate of hospice utilisation at end of life [time frame: 6 months] [designated as safety issue: no] ‐ examine differences in rates of hospice utilisation between study arms

  • Length of stay in hospice [time frame: 6 months] [designated as safety issue: no] ‐ examine differences in hospice length of stay between stud

Starting date

May 2016

Contact information

Jennifer Temel, MD

Massachusetts General Hospital

Bosten, Massachusetts, United States, 02115

E‐Mail: [email protected]

Notes

Last updated on ClinicalTrials.gov on 15 April 2017: "This study is currently recruiting participants."

NCT02853474

Trial name or title

Early palliative care in patients with metastatic upper gastrointestinal cancers treated with first‐line chemotherapy (EPIC‐1511)

Methods

RCT

Participants

Inclusion criteria:

  • Patients with an upper gastrointestinal metastatic cancer: pancreatic, biliary tract, or gastric (including junctional Siewert 2 and 3 cancers) cancers. NB: Gastroesophageal junctional cancers with dysphagia and/or gastric/gastroesophageal cancers with unknown or positive HER2 status are not eligible

  • Patients planning to be treated with first‐line chemotherapy for metastatic disease

  • Age ≥ 18 years

  • Life expectancy ≥ 1 month

  • Performance status (ECOG) ≤ 2

  • Good understanding of French language

  • Signed and dated informed consent

  • Patients covered by government health insurance

Exclusion criteria:

  • Locally advanced cancer

  • Junctional Siewert 1 gastroesophageal cancer

  • Gastric or junctional gastroesophageal cancer with dysphagia

  • Gastric or junctional gastroesophageal cancer with unknown or positive HER2 status (IHC: +++ or IHC ++ and FISH/SISH +)

  • Direct bilirubin > 2 ULN

Target sample size is N = 558

Interventions

Sham comparator: Arm A: chemotherapy alone. The medical oncologists (or gastroenterologist physicians) are in charge of the participant for chemotherapy administration, and for management of symptoms related to the disease and/or the treatment, in accordance with professional practices. If needed (at any time), a palliative consultation visit could be performed. Interventions include the EORTC‐QLQ‐C30 questionnaire for assessment of quality of life and HADS score for anxiety and depression assessment

Experimental: Arm B: chemotherapy + early palliative care. Standard oncology care as for arm A plus early palliative consultation visits. Interventions include EORTC‐QLQ‐C30 questionnaire for assessment of quality of life and HADS score for anxiety and depression assessment. Early palliative care visits: A palliative consultation visit is a visit done by a palliative care physician. Any visits done by other professionals ARE NOT palliative consultation visits. Five palliative consultation visits are scheduled in this arm

Outcomes

Primary outcome measure:

  • Overall survival (as intent‐to treat analysis) [time frame: average of 1 year] [designated as safety issue: no] ‐ overall survival is defined as time between date of randomisation and date of death, whatever the cause

Secondary outcome measures:

  • Overall survival (per‐protocol analysis) [time frame: average of 1 year] [designated as safety issue: no] ‐ overall survival curves in per‐protocol analysis will be given

  • One‐year survival rate (intent‐to‐treat and per‐protocol analyses) [time frame: 1 year] [designated as safety issue: no] ‐ 1‐year survival rates with 95% confidence interval in both intent‐to‐treat and per‐protocol analyses

  • Quality of life [time frame: every 8 weeks until participant withdrawal from the study (during an average of 1 year)] designated as safety issue: no] ‐ quality of life is assessed with the EORTC QLQ‐C30 questionnaire at baseline and, after inclusion, every 8 weeks until participant withdrawal from the study

  • Depression assessed with HADS score [time frame: every 8 weeks during 24 weeks] [designated as safety issue: no] ‐ depression is assessed with the HADS (Hospital Anxiety and Depression Scale) at baseline and, after inclusion, every 8 weeks during 24 weeks. TUDD (time until definitive deterioration) [time frame: average of 1 year] [designated as safety issue: no]

  • TUDD for quality of life scores was defined as the time from randomisation to the first observation of definitive deterioration of EORTC QLQ‐C30 score, or death.

  • Presence or lack of advanced directives [time frame: through study completion, an average of 1 year] [designated as safety issue: no] ‐ number of participants for whom advanced directives are written in their medical records will be recorded

  • Questionnaire on "content of palliative care visits" [time frame: during the 6 first months after randomisation] [designated as safety issue: no] ‐ a palliative care visit is a visit done by a palliative care physician. Any type of visit done by other professionals (i.e. dieticians, nurses, social workers, psychologists, pain specialists, etc.) IS NOT a palliative care visit. In Arm B (interventional arm), the content of each palliative care visit will be described by the palliative care physician at the end of the visit, by filling in a specific checklist built by an ad hoc working group of palliative care physicians

  • Number of participants treated with chemotherapy [time frame: 30 days before death of the participant] [designated as safety issue: no] ‐ number of participants treated with chemotherapy in their 30 last days before death will be recorded

Starting date

August 2016

Contact information

Ariette Da Silva, MD

Antoine Adenis, MD, PhD

Centre Oscar Lambret

3 Rue Frédéric Combemale, 59000 Lille, France

E‐Mail: a‐dasilva@o‐lambret.fr

Notes

Last updated on ClinicalTrials.gov on 10 January 2017: "This study is currently recruiting participants."

BPI: Brief Pain Inventory

CES‐D: Center for Epidemiological Studies‐Depression Scale

ECOG: Eastern Cooperative Oncology Group Performance Status

EORTC QLQ‐C30: European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire

ESAS: Edmonton Symptom Assessment System

FACIT‐Pal: Functional Assessment of Chronic Illness Therapy ‐ Palliative Care

FACIT‐Sp: Functional Assessment of Chronic Illness Therapy ‐ Spirituality

FACT‐Breast: Functional Assessment of Cancer Therapy ‐ Breast

FACT‐E: Functional Assessment of Cancer Therapy ‐ Esophagus

FACT‐G: Functional Assessment of Cancer Therapy ‐ General

FACT‐Hep: Functional Assessment of Cancer Therapy ‐ Hepatobiliary

FACT‐H&N: Functional Assessment of Cancer Therapy ‐ Head and Neck

FACT‐Leu: Functional Assessment of Cancer Therapy – Leukemia

FAMCARE: Family Satisfaction with Advanced Cancer Care Questionnaire

HADS: Hospital Anxiety and Depression Scale

NCCN: National Comprehensive Cancer Network

NSCLC: non‐small cell lung cancer

PCI: palliative care intervention

PHQ‐9: Patient Health Questionnaire

RCT: randomised controlled trial

SSCT: specialist symptom control treatment

SF‐36: Short Form (36) Health Survey

Data and analyses

Open in table viewer
Comparison 1. Early palliative care vs standard oncological care

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Health‐related quality of life Show forest plot

7

1028

Std. Mean Difference (Random, 95% CI)

0.27 [0.15, 0.38]

Analysis 1.1

Comparison 1 Early palliative care vs standard oncological care, Outcome 1 Health‐related quality of life.

Comparison 1 Early palliative care vs standard oncological care, Outcome 1 Health‐related quality of life.

1.1 Co‐ordinated care model

3

485

Std. Mean Difference (Random, 95% CI)

0.21 [0.03, 0.39]

1.2 Integrated care model

4

543

Std. Mean Difference (Random, 95% CI)

0.31 [0.15, 0.46]

2 Depression Show forest plot

5

762

Std. Mean Difference (Random, 95% CI)

‐0.11 [‐0.26, 0.03]

Analysis 1.2

Comparison 1 Early palliative care vs standard oncological care, Outcome 2 Depression.

Comparison 1 Early palliative care vs standard oncological care, Outcome 2 Depression.

2.1 Co‐ordinated care model

3

526

Std. Mean Difference (Random, 95% CI)

‐0.06 [‐0.23, 0.12]

2.2 Integrated care model

2

236

Std. Mean Difference (Random, 95% CI)

‐0.24 [‐0.50, 0.02]

3 Survival Show forest plot

4

800

Hazard Ratio (Random, 95% CI)

0.85 [0.56, 1.28]

Analysis 1.3

Comparison 1 Early palliative care vs standard oncological care, Outcome 3 Survival.

Comparison 1 Early palliative care vs standard oncological care, Outcome 3 Survival.

4 Symptom intensity Show forest plot

7

1054

Std. Mean Difference (Random, 95% CI)

‐0.23 [‐0.35, ‐0.10]

Analysis 1.4

Comparison 1 Early palliative care vs standard oncological care, Outcome 4 Symptom intensity.

Comparison 1 Early palliative care vs standard oncological care, Outcome 4 Symptom intensity.

4.1 Co‐ordinated care model

3

492

Std. Mean Difference (Random, 95% CI)

‐0.23 [‐0.41, ‐0.04]

4.2 Integrated care model

4

562

Std. Mean Difference (Random, 95% CI)

‐0.23 [‐0.43, ‐0.04]

5 Health‐related quality of life (sensitivity analysis for study design including RCTs only) Show forest plot

5

696

Std. Mean Difference (Random, 95% CI)

0.29 [0.14, 0.44]

Analysis 1.5

Comparison 1 Early palliative care vs standard oncological care, Outcome 5 Health‐related quality of life (sensitivity analysis for study design including RCTs only).

Comparison 1 Early palliative care vs standard oncological care, Outcome 5 Health‐related quality of life (sensitivity analysis for study design including RCTs only).

6 Symptom intensity (sensitivity analysis for study design including RCTs only) Show forest plot

5

696

Std. Mean Difference (Random, 95% CI)

‐0.28 [‐0.43, ‐0.13]

Analysis 1.6

Comparison 1 Early palliative care vs standard oncological care, Outcome 6 Symptom intensity (sensitivity analysis for study design including RCTs only).

Comparison 1 Early palliative care vs standard oncological care, Outcome 6 Symptom intensity (sensitivity analysis for study design including RCTs only).

Screening process diagram (as recommended by the PRISMA statement).
Figuras y tablas -
Figure 1

Screening process diagram (as recommended by the PRISMA statement).

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

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

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

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

Forest plot of comparison: 1 Health‐related quality of life, outcome: 1.1 Health‐related quality of life.
Figuras y tablas -
Figure 4

Forest plot of comparison: 1 Health‐related quality of life, outcome: 1.1 Health‐related quality of life.

Forest plot of comparison: 1 Early palliative care vs TAU, outcome: 1.2 Survival.
Figuras y tablas -
Figure 5

Forest plot of comparison: 1 Early palliative care vs TAU, outcome: 1.2 Survival.

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.2 Depression.
Figuras y tablas -
Figure 6

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.2 Depression.

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.4 Symptom intensity.
Figuras y tablas -
Figure 7

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.4 Symptom intensity.

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.5 Health‐related quality of life (sensitivity analysis for study design including RCTs only).
Figuras y tablas -
Figure 8

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.5 Health‐related quality of life (sensitivity analysis for study design including RCTs only).

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.6 Symptom intensity (sensitivity analysis for study design including RCTs only).
Figuras y tablas -
Figure 9

Forest plot of comparison: 1 Early palliative care vs standard oncological care, outcome: 1.6 Symptom intensity (sensitivity analysis for study design including RCTs only).

Comparison 1 Early palliative care vs standard oncological care, Outcome 1 Health‐related quality of life.
Figuras y tablas -
Analysis 1.1

Comparison 1 Early palliative care vs standard oncological care, Outcome 1 Health‐related quality of life.

Comparison 1 Early palliative care vs standard oncological care, Outcome 2 Depression.
Figuras y tablas -
Analysis 1.2

Comparison 1 Early palliative care vs standard oncological care, Outcome 2 Depression.

Comparison 1 Early palliative care vs standard oncological care, Outcome 3 Survival.
Figuras y tablas -
Analysis 1.3

Comparison 1 Early palliative care vs standard oncological care, Outcome 3 Survival.

Comparison 1 Early palliative care vs standard oncological care, Outcome 4 Symptom intensity.
Figuras y tablas -
Analysis 1.4

Comparison 1 Early palliative care vs standard oncological care, Outcome 4 Symptom intensity.

Comparison 1 Early palliative care vs standard oncological care, Outcome 5 Health‐related quality of life (sensitivity analysis for study design including RCTs only).
Figuras y tablas -
Analysis 1.5

Comparison 1 Early palliative care vs standard oncological care, Outcome 5 Health‐related quality of life (sensitivity analysis for study design including RCTs only).

Comparison 1 Early palliative care vs standard oncological care, Outcome 6 Symptom intensity (sensitivity analysis for study design including RCTs only).
Figuras y tablas -
Analysis 1.6

Comparison 1 Early palliative care vs standard oncological care, Outcome 6 Symptom intensity (sensitivity analysis for study design including RCTs only).

Summary of findings for the main comparison. Early palliative care for adults with advanced cancer

Clinical question: Should early palliative care be preferred over treatment as usual for improving health‐related quality of life, depression, and symptom intensity in patients with advanced cancer?

Patient or population: patients with advanced cancer

Settings: mainly outpatient care in Australia, Canada, Italy, and the USA
Intervention: early palliative care

Comparison: treatment as usual

Follow‐up: at 12 weeks or mean difference in repeated measurement results for longitudinal designs

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(studies)

Certainty of the evidence
(GRADE)

Comments

Risk with treatment as usual

Risk with early palliative care

Health‐related quality of life (HRQOL), SD units: measured on FACIT‐Pal, TOI of FACT‐Hep, TOI of FACT‐L, FACT‐G, McGill Quality of Life, FACIT‐Sp. Higher scores indicate better HRQOL. Follow‐up: range 12 weeks to 52 weeks

HRQOL score improved on average 0.27 (95% CI 0.15 to 0.38) SDs more in early palliative care participants than in control participants

1028
(7 RCTs)

⊕⊕⊝⊝
LOW1,2,3

By conventional criteria, an SMD of 0.2 represents a small effect, 0.5 a moderate effect, and 0.8 a large effect (Cohen 1988)

Health‐related quality of life (HRQOL), natural units: measured on FACT‐G (from 0 to 108)

Baseline control group mean score at 70.5 pointsa

HRQOL score improved on average 4.59 (95% CI 2.55 to 6.46) points more in early palliative care participants than in control participants

1028
(7 RCTs)

⊕⊕⊝⊝
LOW1,2,3

Calculated by transforming all scales to the FACT‐G in which the minimal clinically important difference is approximately 5 and the SD in the cancer validation sample was 17.0 (Brucker 2005)

Survival: estimated with the unadjusted death hazard ratio

Study populationb

HR 0.85, 95% CI 0.56 to 1.28

800
(4 RCTs)

⊕⊝⊝⊝
VERY LOW1,4,5,6

61 per 100

56 per 100 (41‐71)

Depression, SD units: measured on CES‐D, HADS‐D, PHQ‐9. Higher scores indicate higher depressive symptom load. Follow‐up: range 12 weeks to 52 weeks

Depression score improved on average ‐0.11 (95% CI ‐0.26 to 0.03) SDs more in early palliative care participants than in control participants

762
(5 RCTs)

⊕⊝⊝⊝

VERY LOW1,2,4

By conventional criteria, an SMD of 0.2 represents a small effect, 0.5 a moderate effect, and 0.8 a large effect (Cohen 1988)

Depression, natural units: measured on CES‐D (from 0 to 60). Higher scores indicate higher depressive symptom load

Baseline control group mean score at 13.8 pointsc

Depressive symptoms score improved on average ‐0.98 (95% CI ‐2.31 to 0.27) points more in early palliative care participants than in control participants

762
(5 RCTs)

⊕⊝⊝⊝

VERY LOW1,2,4

Calculated by transforming all scales to CES‐D

and applying an SD of 8.9 from baseline control group score in Bakitas 2009

Symptom intensity, SD units: measured on ESAS, QUAL‐E Symptom Impact Subscale, SDS, RSC, LCS of FACT‐L, HCS of FACT‐Hep. Higher scores indicate higher symptom intensity. Follow‐up: range 12 weeks to 52 weeks

Symptom intensity score improved on average ‐0.23 (95% CI ‐0.35 to ‐0.1) SDs more in early palliative care participants than in control participants

1054
(7 RCTs)

⊕⊕⊝⊝
LOW1,2,3

By conventional criteria, an SMD of 0.2 represents a small effect, 0.5 a moderate effect, and 0.8 a large effect (Cohen 1988)

Symptom intensity, natural units: measured on ESAS (from 0 to 900). Follow‐up: range 12 weeks to 52 weeks

Baseline control group mean score at 286.3 pointsc

Symptom intensity symptoms score improved on average ‐35.4 (95% CI ‐53.9 to ‐15.4) points more in early palliative care participants than in control participants

1054
(7 RCTs)

⊕⊕⊝⊝
LOW1,2,3

Calculated by transforming all scales to the ESAS and applying an SD of 154.0 from baseline control group score in Bakitas 2009

Adverse events

See comment

See comment

Not estimable

1614
(7 RCTs)

See comment

Most often, study authors did not address assessment or findings on adverse events in their study publications. However, on request, authors of 6 studies described the tolerability of early palliative care as very good. A single study mentioned adverse events only in the early palliative care group, i.e. higher percentage of participants with severe scores for pain and poor appetite along with higher level of unmet needs (Tattersall 2014)

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

aApproximate average of baseline control group FACT‐G scores across 4 included studies (Bakitas 2009; Bakitas 2015; Maltoni 2016; Temel 2010)

b12‐Month follow‐up control group risk in the largest study reporting on survival (Bakitas 2009)

cBaseline control group CES‐D score in the largest study reporting on depression (Bakitas 2009)

CI: confidence interval; GRADE: Grading of Recommendations Assessment; HR: unadjusted death hazard ratio; SD: standard deviation; SMD: standardised mean difference

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.

1We downgraded 2 points owing to very serious limitations in study quality (high risk of bias across studies)

2We decided against downgrading for indirectness, although 2 studies were conducted exclusively in patients with metastatic pancreatic and advanced lung cancer, respectively (Maltoni 2016; Temel 2010). We decided against downgrading for inconsistency, as we did not detect significant heterogeneity

3We decided against downgrading for imprecision, as the optimal information size (OIS) criterion was met, and the 95% confidence interval around the difference in effect between intervention and control excludes zero

4We downgraded 1 point for imprecision, as the optimal information size (OIS) criterion was met, but the 95% confidence interval around the difference in effect between intervention and control includes zero. The 95% confidence interval fails to exclude harm

5We decided against downgrading for important inconsistency (large I2) because we had downgraded by 3 points already

6We decided against downgrading for indirectness, as only a single study was conducted exclusively in patients with advanced lung cancer (Temel 2010)

Figuras y tablas -
Summary of findings for the main comparison. Early palliative care for adults with advanced cancer
Comparison 1. Early palliative care vs standard oncological care

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Health‐related quality of life Show forest plot

7

1028

Std. Mean Difference (Random, 95% CI)

0.27 [0.15, 0.38]

1.1 Co‐ordinated care model

3

485

Std. Mean Difference (Random, 95% CI)

0.21 [0.03, 0.39]

1.2 Integrated care model

4

543

Std. Mean Difference (Random, 95% CI)

0.31 [0.15, 0.46]

2 Depression Show forest plot

5

762

Std. Mean Difference (Random, 95% CI)

‐0.11 [‐0.26, 0.03]

2.1 Co‐ordinated care model

3

526

Std. Mean Difference (Random, 95% CI)

‐0.06 [‐0.23, 0.12]

2.2 Integrated care model

2

236

Std. Mean Difference (Random, 95% CI)

‐0.24 [‐0.50, 0.02]

3 Survival Show forest plot

4

800

Hazard Ratio (Random, 95% CI)

0.85 [0.56, 1.28]

4 Symptom intensity Show forest plot

7

1054

Std. Mean Difference (Random, 95% CI)

‐0.23 [‐0.35, ‐0.10]

4.1 Co‐ordinated care model

3

492

Std. Mean Difference (Random, 95% CI)

‐0.23 [‐0.41, ‐0.04]

4.2 Integrated care model

4

562

Std. Mean Difference (Random, 95% CI)

‐0.23 [‐0.43, ‐0.04]

5 Health‐related quality of life (sensitivity analysis for study design including RCTs only) Show forest plot

5

696

Std. Mean Difference (Random, 95% CI)

0.29 [0.14, 0.44]

6 Symptom intensity (sensitivity analysis for study design including RCTs only) Show forest plot

5

696

Std. Mean Difference (Random, 95% CI)

‐0.28 [‐0.43, ‐0.13]

Figuras y tablas -
Comparison 1. Early palliative care vs standard oncological care