open access

Vol 72, No 6 (2022)
Guidelines / Expert consensus
Published online: 2022-12-07
Get Citation

Post-treatment follow-up in common solid malignancies: expert panel recommendations

Jacek Jassem1, Anna Kowalczyk1, Aleksander Biesiada23, Renata Duchnowska4, Rafał Dziadziuszko1, Agnieszka Mastalerz-Migas5, Andrzej Kawecki6, Maciej Krzakowski7, Piotr Potemski89, Piotr Rutkowski10, Piotr Wysocki11
·
Nowotwory. Journal of Oncology 2022;72(6):384-407.
Affiliations
  1. Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
  2. Ad Vitam Sp. z o.o. Outpatient Clinic, Skawina, Poland
  3. Siloe Home Hospice, Krakow, Poland
  4. Department of Oncology, Military Institute of Medicine, Warsaw, Poland
  5. Department of Family Medicine, Wroclaw Medical University, Wroclaw, Poland
  6. Department of Head and Neck Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
  7. Department of Lung Cancer and Thoracic Tumours, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
  8. Department of Chemotherapy, Medical University of Lodz, Lodz, Poland
  9. Mikołaj Kopernik Voivodeship Multi-Speciality Centre for Oncology and Traumatology in Lodz, Lodz, Poland
  10. Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
  11. Department of Oncology, Collegium Medicum, Jagiellonian University, Krakow, Poland

open access

Vol 72, No 6 (2022)
Guidelines and recommendations
Published online: 2022-12-07

Abstract

Post-treatment follow-up is an essential component of comprehensive cancer care. Determining optimal follow-up schedules is crucial on clinical, organisational and economic grounds. Owing to the scarcity of prospective clinical follow-up trials, most recommendations are based on retrospective studies and expert opinions. In 2014, the first post­-treatment follow-up recommendations in the most common solid malignancies was published by Polish oncology and family medicine experts. In this article, we present an update of this document that takes into account the current literature and the quality of the available scientific evidence.

Abstract

Post-treatment follow-up is an essential component of comprehensive cancer care. Determining optimal follow-up schedules is crucial on clinical, organisational and economic grounds. Owing to the scarcity of prospective clinical follow-up trials, most recommendations are based on retrospective studies and expert opinions. In 2014, the first post­-treatment follow-up recommendations in the most common solid malignancies was published by Polish oncology and family medicine experts. In this article, we present an update of this document that takes into account the current literature and the quality of the available scientific evidence.

Get Citation

Keywords

cancer; post-treatment follow-up; recommendations

About this article
Title

Post-treatment follow-up in common solid malignancies: expert panel recommendations

Journal

Nowotwory. Journal of Oncology

Issue

Vol 72, No 6 (2022)

Article type

Guidelines / Expert consensus

Pages

384-407

Published online

2022-12-07

Page views

3545

Article views/downloads

322

DOI

10.5603/NJO.a2022.0058

Bibliographic record

Nowotwory. Journal of Oncology 2022;72(6):384-407.

Keywords

cancer
post-treatment follow-up
recommendations

Authors

Jacek Jassem
Anna Kowalczyk
Aleksander Biesiada
Renata Duchnowska
Rafał Dziadziuszko
Agnieszka Mastalerz-Migas
Andrzej Kawecki
Maciej Krzakowski
Piotr Potemski
Piotr Rutkowski
Piotr Wysocki

References (182)
  1. Jassem J, Duchnowska R, Kawecki A, et al. Badania kontrolne po leczeniu w najczęstszych nowotworach litych u dorosłych. Nowotwory. Journal of Oncology. 2014; 64(5): 415–435.
  2. Appraisal of Guidelines for Research and Evaluation (AGREE). http://www.agreetrust.org (30.08.2022).
  3. Kearney PL, Watkins JM, Shirai K, et al. Salvage Resection for Isolated Local and/or Regional Failure of Head/Neck Cancer Following Definitive Concurrent Chemoradiotherapy Case Series and Review of the Literature. Mcgill J Med. 2011; 13(2): 29.
  4. Lee DH, Roh JL, Baek S, et al. Second cancer incidence, risk factor, and specific mortality in head and neck squamous cell carcinoma. Otolaryngol Head Neck Surg. 2013; 149(4): 579–586.
  5. Jassem J. Tobacco smoking after diagnosis of cancer: clinical aspects. Transl Lung Cancer Res. 2019; 8(Suppl 1): S50–S58.
  6. Bała MM, Cedzyńska M, Balwicki Ł, et al. Wytyczne leczenia uzależnienia od nikotyny. Med Prakt. 2022: 22–40.
  7. Goodwin WJ. Salvage surgery for patients with recurrent squamous cell carcinoma of the upper aerodigestive tract: when do the ends justify the means? Laryngoscope. 2000; 110(3 Pt 2 Suppl 93): 1–18.
  8. Oral Care Study Group. Evidence-based management strategies for oral complication from cancer treatment. MASCC/ISOO; 2011. www.mascc.org/isoo (30.08.2022).
  9. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Head and Neck Cancers. Wersja 2.2022. www.nccn.org (30.08.2022).
  10. Manikantan K, Khode S, Dwivedi RC, et al. Making sense of post-treatment surveillance in head and neck cancer: when and what of follow-up. Cancer Treat Rev. 2009; 35(8): 744–753.
  11. Simo R, Homer J. Follow-up of Head and Neck Cancers. Head and neck cancer: multidisciplinary management guidelines. ENT-UK, London 2011: 362–366.
  12. Louis D, Perry A, Wesseling P, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol. 2021; 23(8): 1231–1251.
  13. Monroe CL, Travers S, Woldu HG, et al. Does Surveillance-Detected Disease Progression Yield Superior Patient Outcomes in High-Grade Glioma? World Neurosurg. 2020; 135: e410–e417.
  14. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Central Nervous System Cancers. Wersja 1.2022. www.nccn.org (30.08.2022).
  15. Jo J, van den Bent MJ, Nabors B, et al. Surveillance imaging frequency in adult patients with lower-grade (WHO Grade 2 and 3) gliomas. Neuro Oncol. 2022; 24(7): 1035–1047.
  16. Abdalla G, Hammam A, Anjari M, et al. Glioma surveillance imaging: current strategies, shortcomings, challenges and outlook. BJR Open. 2020; 2(1): 20200009.
  17. Geßler F, Dützmann S, Quick J, et al. Is postoperative imaging mandatory after meningioma removal? Results of a prospective study. PLoS One. 2015; 10(4): e0124534.
  18. Islim AI, Kolamunnage-Dona R, Mohan M, et al. A prognostic model to personalize monitoring regimes for patients with incidental asymptomatic meningiomas. Neuro Oncol. 2020; 22(2): 278–289.
  19. Colt HG, Murgu SD, Korst RJ, et al. Follow-up and surveillance of the patient with lung cancer after curative-intent therapy: Diagnosis and management of lung cancer, 3rd ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2013; 143(5 Suppl): e437S–e454S.
  20. Lou F, Huang J, Sima CS, et al. Patterns of recurrence and second primary lung cancer in early-stage lung cancer survivors followed with routine computed tomography surveillance. J Thorac Cardiovasc Surg. 2013; 145(1): 75–81; discussion 81.
  21. Schmidt-Hansen M, Baldwin DR, Hasler E. What is the most effective follow-up model for lung cancer patients? A systematic review. J Thorac Oncol. 2012; 7(5): 821–824.
  22. Aupérin A, Le Péchoux C, Rolland E, et al. Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer. J Clin Oncol. 2010; 28(13): 2181–2190.
  23. Nakamura R, Kurishima K, Kobayashi N, et al. Postoperative follow-up for patients with non-small cell lung cancer. Onkologie. 2010; 33(1-2): 14–18.
  24. Crabtree TD, Puri V, Chen SB, et al. Does the method of radiologic surveillance affect survival after resection of stage I non-small cell lung cancer? J Thorac Cardiovasc Surg. 2015; 149(1): 45–52, 53.e1.
  25. Srikantharajah D, Ghuman A, Nagendran M, et al. Is computed tomography follow-up of patients after lobectomy for non-small cell lung cancer of benefit in terms of survival? Interact Cardiovasc Thorac Surg. 2012; 15(5): 893–898.
  26. Westeel V, Foucher P, Scherpereel A, et al. Chest CT scan plus x-ray versus chest x-ray for the follow-up of completely resected non-small-cell lung cancer (IFCT-0302): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2022; 23(9): 1180–1188.
  27. McMurry T, Stukenborg G, Kessler L, et al. More Frequent Surveillance Following Lung Cancer Resection Is Not Associated With Improved Survival. Ann Surg. 2018; 268(4): 632–639.
  28. Denis F, Lethrosne C, Pourel N, et al. Randomized Trial Comparing a Web-Mediated Follow-up With Routine Surveillance in Lung Cancer Patients. J Natl Cancer Inst. 2017; 109(9).
  29. Sugiyama T, Hirose T, Hosaka T, et al. Effectiveness of intensive follow-up after response in patients with small cell lung cancer. Lung Cancer. 2008; 59(2): 255–261.
  30. Takahashi T, Yamanaka T, Seto T, et al. Prophylactic cranial irradiation versus observation in patients with extensive-disease small-cell lung cancer: a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2017; 18(5): 663–671.
  31. Baudin E, Caplin M, Garcia-Carbonero R, et al. ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Lung and thymic carcinoids: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021; 32(4): 439–451.
  32. Sinha S, Swift AJ, Kamil MA, et al. The role of imaging in malignant pleural mesothelioma: an update after the 2018 BTS guidelines. Clin Radiol. 2020; 75(6): 423–432.
  33. Falkson CB, Bezjak A, Darling G, et al. Lung Cancer Disease Site Group of Cancer Care Ontario's Program in Evidence-Based Care. The management of thymoma: a systematic review and practice guideline. J Thorac Oncol. 2009; 4(7): 911–919.
  34. Jeffery M, Hickey BE, Hider PN. Follow-up strategies for patients treated for non-metastatic colorectal cancer. Cochrane Database Syst Rev. 2019; 9(9): CD002200.
  35. Primrose JN, Perera R, Gray A, et al. FACS Trial Investigators. Effect of 3 to 5 years of scheduled CEA and CT follow-up to detect recurrence of colorectal cancer: the FACS randomized clinical trial. JAMA. 2014; 311(3): 263–270.
  36. Argilés G, Tabernero J, Labianca R, et al. ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Localised colon cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020; 31(10): 1291–1305.
  37. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Colon cancer. Wersja 3.2022. www.nccn.org (30.08.2022).
  38. Meyerhardt JA, Mangu PB, Flynn PJ, et al. American Society of Clinical Oncology. Follow-up care, surveillance protocol, and secondary prevention measures for survivors of colorectal cancer: American Society of Clinical Oncology clinical practice guideline endorsement. J Clin Oncol. 2013; 31(35): 4465–4470.
  39. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Rectal cancer. Wersja 1.2022. www.nccn.org (30.08.2022).
  40. Glynne-Jones R, Wyrwicz L, Tiret E, et al. ESMO Guidelines Committee. Rectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017; 28(suppl_4): iv22–iv40.
  41. Khatcheressian JL, Hurley P, Bantug E, et al. American Society of Clinical Oncology. Breast cancer follow-up and management after primary treatment: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2013; 31(7): 961–965.
  42. Senkus E, Kyriakides S, Penault-Llorca F, et al. ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013; 24(supl. 6): vi7–vi23.
  43. Rojas MP, Telaro E, Russo A, et al. Follow-up strategies for women treated for early breast cancer. Cochrane Database Syst Rev. 2000(4): CD001768.
  44. Impact of follow-up testing on survival and health-related quality of life in breast cancer patients. A multicenter randomized controlled trial. The GIVIO Investigators. JAMA. 1994; 271(20): 1587–1592.
  45. Goldhirsch A, Winer EP, Coates AS, et al. Panel members. Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol. 2013; 24(9): 2206–2223.
  46. Grunfeld E, Levine MN, Julian JA, et al. Randomized trial of long-term follow-up for early-stage breast cancer: a comparison of family physician versus specialist care. J Clin Oncol. 2006; 24(6): 848–855.
  47. Jassem J, Krzakowski M, Bobek-Billewicz B, et al. Rak piersi. Onk Prakt Klin Edu. 2020; 6: 297–532.
  48. Lu W, de Bock GH, Schaapveld M, et al. The value of routine physical examination in the follow up of women with a history of early breast cancer. Eur J Cancer. 2011; 47(5): 676–682.
  49. Lash TL, Fox MP, Silliman RA. Reduced mortality rate associated with annual mammograms after breast cancer therapy. Breast J. 2006; 12(1): 2–6.
  50. Berg WA, Zhang Z, Cormack JB, et al. ACRIN 6666 Investigators. Detection of breast cancer with addition of annual screening ultrasound or a single screening MRI to mammography in women with elevated breast cancer risk. JAMA. 2012; 307(13): 1394–1404.
  51. Quinn EM, Coveney AP, Redmond HP. Use of magnetic resonance imaging in detection of breast cancer recurrence: a systematic review. Ann Surg Oncol. 2012; 19(9): 3035–3041.
  52. Barnsley GP, Grunfeld E, Coyle D, et al. Surveillance mammography following the treatment of primary breast cancer with breast reconstruction: a systematic review. Plast Reconstr Surg. 2007; 120(5): 1125–1132.
  53. Smid M, Wang Y, Zhang Yi, et al. Subtypes of breast cancer show preferential site of relapse. Cancer Res. 2008; 68(9): 3108–3114.
  54. Khan QJ, Reddy PS, Kimler BF, et al. Effect of vitamin D supplementation on serum 25-hydroxy vitamin D levels, joint pain, and fatigue in women starting adjuvant letrozole treatment for breast cancer. Breast Cancer Res Treat. 2010; 119(1): 111–118.
  55. Rastelli AL, Taylor ME, Gao F, et al. Vitamin D and aromatase inhibitor-induced musculoskeletal symptoms (AIMSS): a phase II, double-blind, placebo-controlled, randomized trial. Breast Cancer Res Treat. 2011; 129(1): 107–116.
  56. Głuszko P, Tłustochowicz W, Korkosz M. Choroby metaboliczne kości. In: Gajewski P, Korkosz M. ed. Interna Szczeklika 2021. Medycyna Praktyczna, Kraków 2021: 2165–2175.
  57. Kroenke CH, Chen WY, Rosner B, et al. Weight, weight gain, and survival after breast cancer diagnosis. J Clin Oncol. 2005; 23(7): 1370–1378.
  58. Holmes M, Chen WY, Feskanich D, et al. Physical Activity and Survival After Breast Cancer Diagnosis. JAMA. 2005; 293(20): 2479.
  59. Holmberg L, Anderson H. HABITS (hormonal replacement therapy after breast cancer—is it safe?), a randomised comparison: trial stopped. Lancet. 2004; 363(9407): 453–455.
  60. Zhao S, Chlebowski RT, Anderson GL, et al. Sex hormone associations with breast cancer risk and the mediation of randomized trial postmenopausal hormone therapy effects. Breast Cancer Res. 2014; 16(2): R30.
  61. Moegele M, Buchholz S, Seitz S, et al. Vaginal Estrogen Therapy for Patients with Breast Cancer. Geburtshilfe Frauenheilkd. 2013; 73(10): 1017–1022.
  62. Le Ray I, Dell'Aniello S, Bonnetain F, et al. Local estrogen therapy and risk of breast cancer recurrence among hormone-treated patients: a nested case-control study. Breast Cancer Res Treat. 2012; 135(2): 603–609.
  63. Sánchez-Rovira P, Hirschberg AL, Gil-Gil M, et al. A Phase II Prospective, Randomized, Double-Blind, Placebo-Controlled and Multicenter Clinical Trial to Assess the Safety of 0.005% Estriol Vaginal Gel in Hormone Receptor-Positive Postmenopausal Women with Early Stage Breast Cancer in Treatment with Aromatase Inhibitor in the Adjuvant Setting. Oncologist. 2020; 25(12): e1846–e1854.
  64. Melisko ME, Goldman ME, Hwang J, et al. Vaginal Testosterone Cream vs Estradiol Vaginal Ring for Vaginal Dryness or Decreased Libido in Women Receiving Aromatase Inhibitors for Early-Stage Breast Cancer: A Randomized Clinical Trial. JAMA Oncol. 2017; 3(3): 313–319.
  65. Salani R, Khanna N, Frimer M, et al. An update on post-treatment surveillance and diagnosis of recurrence in women with gynecologic malignancies: Society of Gynecologic Oncology (SGO) recommendations. Gynecol Oncol. 2017; 146(1): 3–10.
  66. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Uterine Neoplasms. Wersja 1.2022. www.nccn.org (30.08.2022).
  67. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Cervical cancer. Wersja 1.2022. www.nccn.org (30.08.2022).
  68. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Ovarian Cancer/Fallopian Tube Cancer/Primary Peritoneal Cancer. Wersja 4.2022. www.nccn.org (30.08.2022).
  69. Marth C, Landoni F, Mahner S, et al. ESMO Guidelines Committee. Cervical cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017; 28(suppl_4): iv72–iv83.
  70. Ledermann JA, Raja FA, Fotopoulou C, et al. ESMO Guidelines Working Group. Newly diagnosed and relapsed epithelial ovarian carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013; 24 Suppl 6: vi24–vi32.
  71. Colombo N, Preti E, Landoni F, et al. ESMO Guidelines Working Group. Endometrial cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2013; 24 Suppl 6: vi33–vi38.
  72. Newton C, Nordin A, Rolland P, et al. British Gynaecological Cancer Society recommendations and guidance on patient-initiated follow-up (PIFU). Int J Gynecol Cancer. 2020; 30(5): 695–700.
  73. Fung-Kee-Fung M, Dodge J, Elit L, et al. Cancer Care Ontario Program in Evidence-based Care Gynecology Cancer Disease Site Group. Follow-up after primary therapy for endometrial cancer: a systematic review. Gynecol Oncol. 2006; 101(3): 520–529.
  74. Zola P, Ciccone G, Piovano E, et al. Intensive versus minimalist follow-up in patients treated for endometrial cancer: A multicentric randomized controlled trial (The TOTEM study—NCT00916708). J Clinl Oncol. 2021; 39(15_suppl): 5506–5506.
  75. Elit L, Fyles AW, Oliver TK, et al. Follow-Up for Women after Treatment for Cervical Cancer. Curr Oncol. 2010; 17(3): 65–69.
  76. Zanagnolo V, Ming L, Gadducci A, et al. Surveillance procedures for patients with cervical carcinoma: a review of the literature. Int J Gynecol Cancer. 2009; 19(2): 194–201.
  77. Brooks RA, Rader JS, Dehdashti F, et al. Surveillance FDG-PET detection of asymptomatic recurrences in patients with cervical cancer. Gynecol Oncol. 2009; 112(1): 104–109.
  78. Gonzalez Bosquet J, Magrina JF, Gaffey TA, et al. Long-term survival and disease recurrence in patients with primary squamous cell carcinoma of the vulva. Gynecol Oncol. 2005; 97(3): 828–833.
  79. Tantipalakorn C, Robertson G, Marsden DE, et al. Outcome and patterns of recurrence for International Federation of Gynecology and Obstetrics (FIGO) stages I and II squamous cell vulvar cancer. Obstet Gynecol. 2009; 113(4): 895–901.
  80. Gadducci A, Fuso L, Cosio S, et al. Are surveillance procedures of clinical benefit for patients treated for ovarian cancer?: A retrospective Italian multicentric study. Int J Gynecol Cancer. 2009; 19(3): 367–374.
  81. Rustin GJ, van de, Griffin CL, et al. Early versus delayed treatment of relapsed ovarian cancer (MRC OV05/EORTC 55955): a randomised trial. Lancet. 2010; 376(9747): 1155–1163.
  82. Scaletta G, Plotti F, Luvero D, et al. The role of novel biomarker HE4 in the diagnosis, prognosis and follow-up of ovarian cancer: a systematic review. Expert Rev Anticancer Ther. 2017; 17(9): 827–839.
  83. Bois Adu, Reuss A, Pujade-Lauraine E, et al. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: A combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials. Cancer. 2009; 115(6): 1234–1244.
  84. du Bois A, Ewald-Riegler N, de Gregorio N, et al. Arbeitsgmeinschaft Gynäkologische Onkologie (AGO) Study Group. Borderline tumours of the ovary: A cohort study of the Arbeitsgmeinschaft Gynäkologische Onkologie (AGO) Study Group. Eur J Cancer. 2013; 49(8): 1905–1914.
  85. Vasconcelos I, de Sousa Mendes M. Conservative surgery in ovarian borderline tumours: a meta-analysis with emphasis on recurrence risk. Eur J Cancer. 2015; 51(5): 620–631.
  86. Zanetta G, Rota S, Lissoni A, et al. Ultrasound, physical examination, and CA 125 measurement for the detection of recurrence after conservative surgery for early borderline ovarian tumors. Gynecol Oncol. 2001; 81(1): 63–66.
  87. Kane A, Uzan C, Rey A, et al. Prognostic factors in patients with ovarian serous low malignant potential (borderline) tumors with peritoneal implants. Oncologist. 2009; 14(6): 591–600.
  88. Schumer ST, Cannistra SA. Granulosa cell tumor of the ovary. J Clin Oncol. 2003; 21(6): 1180–1189.
  89. Gershenson DM. Management of ovarian germ cell tumor. J Clin Oncol. 2007; 25: 2938–2943.
  90. Shen S, Lepor H, Yaffee R, et al. Ultrasensitive serum prostate specific antigen nadir accurately predicts the risk of early relapse after radical prostatectomy. J Urol. 2005; 173(3): 777–780.
  91. Eisenberg ML, Davies BJ, Cooperberg MR, et al. Prognostic implications of an undetectable ultrasensitive prostate-specific antigen level after radical prostatectomy. Eur Urol. 2010; 57(4): 622–629.
  92. Cheung R, Roach M, Hanks G, et al. Defining biochemical failure following radiotherapy with or without hormonal therapy in men with clinically localized prostate cancer: recommendations of the RTOG-ASTRO Phoenix Consensus Conference. Int J Radiat Oncol Biol Phys. 2006; 65(4): 965–974.
  93. Stamey T, Kabalin J, Mcneal J, et al. Prostate Specific Antigen in the Diagnosis and Treatment of Adenocarcinoma of the Prostate. II. Radical Prostatectomy Treated Patients. J Urol. 1989; 141(5): 1076–1083.
  94. Partin AW, Pearson JD, Landis PK, et al. Evaluation of serum prostate-specific antigen velocity after radical prostatectomy to distinguish local recurrence from distant metastases. Urology. 1994; 43(5): 649–659.
  95. Ray ME, Thames HD, Levy LB, et al. PSA nadir predicts biochemical and distant failures after external beam radiotherapy for prostate cancer: a multi-institutional analysis. Int J Radiat Oncol Biol Phys. 2006; 64(4): 1140–1150.
  96. Hancock S, Cox R, Bagshaw M. Prostate Specific Antigen After Radiotherapy for Prostate Cancer. J Urol. 1995: 1412–1417.
  97. Oefelein M, Smith N, Carter M, et al. The Incidence of Prostate Cancer Progression with Undetectable Serum Prostate Specific Antigen in a Series of 394 Radical Prostatectomies. J Urol. 1995: 2128–2131.
  98. Sorbellini M, Kattan MW, Snyder ME, et al. A postoperative prognostic nomogram for renal cell carcinoma. J Urol. 2001; 166(1): 63–67.
  99. LAM J, SHVARTS O, LEPPERT J, et al. POSTOPERATIVE SURVEILLANCE PROTOCOL FOR PATIENTS WITH LOCALIZED AND LOCALLY ADVANCED RENAL CELL CARCINOMA BASED ON A VALIDATED PROGNOSTIC NOMOGRAM AND RISK GROUP STRATIFICATION SYSTEM. Journal of Urology. 2005; 174(2): 466–472.
  100. Leibovich BC, Blute ML, Cheville JC, et al. Prediction of progression after radical nephrectomy for patients with clear cell renal cell carcinoma: a stratification tool for prospective clinical trials. Cancer. 2003; 97(7): 1663–1671.
  101. Karakiewicz PI, Briganti A, Chun FKH, et al. Multi-institutional validation of a new renal cancer-specific survival nomogram. J Clin Oncol. 2007; 25(11): 1316–1322.
  102. Bellmunt J, Orsola A, Leow JJ, et al. ESMO Guidelines Working Group. Bladder cancer: ESMO Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2014; 25 Suppl 3: iii40–iii48.
  103. Albers P, Albrecht W, Algaba F, et al. Guidelines on Testicular Cancer. European Urology. 2011; 60: 304–319.
  104. Hakenberg OW, Compérat EM, Minhas S, et al. EAU guidelines on penile cancer: 2014 update. Eur Urol. 2015; 67(1): 142–150.
  105. Francken AB, Hoekstra HJ. Follow-up of melanoma patients: the need for evidence-based protocols. Ann Surg Oncol. 2009; 16(4): 804–805.
  106. Fields RC, Coit DG. Evidence-based follow-up for the patient with melanoma. Surg Oncol Clin N Am. 2011; 20(1): 181–200.
  107. Scally CP, Wong SL. Intensity of follow-up after melanoma surgery. Ann Surg Oncol. 2014; 21(3): 752–757.
  108. Weiss M, Loprinzi CL, Creagan ET, et al. Utility of follow-up tests for detecting recurrent disease in patients with malignant melanomas. JAMA. 1995; 274(21): 1703–1705.
  109. Michielin O, van Akkooi ACJ, Ascierto PA, et al. ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Cutaneous melanoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 2019; 30(12): 1884–1901.
  110. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Melanoma: Cutaneous. Wersja 3.2022. www.nccn.org (30.08.2022).
  111. Francken A, Bastiaannet E, Hoekstra H. Follow-up in patients with localised primary cutaneous melanoma. Lancet Oncol. 2005; 6(8): 608–621.
  112. Francken AB, Shaw HM, Accortt NA, et al. Detection of first relapse in cutaneous melanoma patients: implications for the formulation of evidence-based follow-up guidelines. Ann Surg Oncol. 2007; 14(6): 1924–1933.
  113. Garbe C, Paul A, Kohler-Späth H, et al. Prospective evaluation of a follow-up schedule in cutaneous melanoma patients: recommendations for an effective follow-up strategy. J Clin Oncol. 2003; 21(3): 520–529.
  114. Meyers MO, Yeh JJ, Frank J, et al. Method of detection of initial recurrence of stage II/III cutaneous melanoma: analysis of the utility of follow-up staging. Ann Surg Oncol. 2009; 16(4): 941–947.
  115. Romano E, Scordo M, Dusza SW, et al. Site and timing of first relapse in stage III melanoma patients: implications for follow-up guidelines. J Clin Oncol. 2010; 28(18): 3042–3047.
  116. Rueth NM, Xing Y, Chiang YJ, et al. Is surveillance imaging effective for detecting surgically treatable recurrences in patients with melanoma? A comparative analysis of stage-specific surveillance strategies. Ann Surg. 2014; 259(6): 1215–1222.
  117. Turner RM, Bell KJL, Morton RL, et al. Optimizing the frequency of follow-up visits for patients treated for localized primary cutaneous melanoma. J Clin Oncol. 2011; 29(35): 4641–4646.
  118. Tsao H, Feldman M, Fullerton JE, et al. Early detection of asymptomatic pulmonary melanoma metastases by routine chest radiographs is not associated with improved survival. Arch Dermatol. 2004; 140(1): 67–70.
  119. Autier P, Coebergh JW, Boniol M, et al. Management of melanoma patients: benefit of intense follow-up schedule is not demonstrated. J Clin Oncol. 2003; 21(19): 3707; author reply 3707–8.
  120. Einwachter-Thompson J, MacKie RM. An evidence base for reconsidering current follow-up guidelines for patients with cutaneous melanoma less than 0.5 mm thick at diagnosis. Br J Dermatol. 2008; 159(2): 337–341.
  121. Moncrieff MD, Underwood B, Garioch JJ, et al. The MelFo Study UK: Effects of a Reduced-Frequency, Stage-Adjusted Follow-Up Schedule for Cutaneous Melanoma 1B to 2C Patients After 3-Years. Ann Surg Oncol. 2020; 27(11): 4109–4119.
  122. Deckers EA, Hoekstra-Weebers JE, Damude S, et al. The MELFO Study: A Multicenter, Prospective, Randomized Clinical Trial on the Effects of a Reduced Stage-Adjusted Follow-Up Schedule on Cutaneous Melanoma IB-IIC Patients-Results After 3 Years. Ann Surg Oncol. 2020; 27(5): 1407–1417.
  123. Park TS, Phan GQ, Yang JC, et al. Routine Computer Tomography Imaging for the Detection of Recurrences in High-Risk Melanoma Patients. Ann Surg Oncol. 2017; 24(4): 947–951.
  124. Rutkowski P, Lugowska I. Follow-up in melanoma patients. Memo. 2014; 7(2): 83–86.
  125. Xing Y, Bronstein Y, Ross MI, et al. Contemporary diagnostic imaging modalities for the staging and surveillance of melanoma patients: a meta-analysis. J Natl Cancer Inst. 2011; 103(2): 129–142.
  126. Titus-Ernstoff L, Perry AE, Spencer SK, et al. Multiple primary melanoma: two-year results from a population-based study. Arch Dermatol. 2006; 142(4): 433–438.
  127. Schuurman MS, de Waal AC, Thijs EJM, et al. Risk factors for second primary melanoma among Dutch patients with melanoma. Br J Dermatol. 2017; 176(4): 971–978.
  128. Youlden DR, Youl PH, Soyer HP, et al. Distribution of subsequent primary invasive melanomas following a first primary invasive or in situ melanoma Queensland, Australia, 1982-2010. JAMA Dermatol. 2014; 150(5): 526–534.
  129. Lallas A, Apalla Z, Kyrgidis A, et al. Second primary melanomas in a cohort of 977 melanoma patients within the first 5 years of monitoring. J Am Acad Dermatol. 2020; 82(2): 398–406.
  130. Salama AKS, de Rosa N, Scheri RP, et al. Hazard-rate analysis and patterns of recurrence in early stage melanoma: moving towards a rationally designed surveillance strategy. PLoS One. 2013; 8(3): e57665.
  131. Cool P, Grimer R, Rees R. Surveillance in patients with sarcoma of the extremities. Eur J Surg Oncol. 2005; 31(9): 1020–1024.
  132. Goel A, Christy MEL, Virgo KS, et al. Costs of follow-up after potentially curative treatment for extremity soft-tissue sarcoma. Int J Oncol. 2004; 25(2): 429–435.
  133. Gerrand CH, Billingham LJ, Woll PJ, et al. Follow up after Primary Treatment of Soft Tissue Sarcoma: A Survey of Current Practice in the United Kingdom. Sarcoma. 2007; 2007: 34128.
  134. Casali PG, Blay JY, Abecassis N, et al. ESMO Guidelines Committee, EURACAN and GENTURIS. Electronic address: clinicalguidelines@esmo.org, ESMO Guidelines Committee, EURACAN and GENTURIS. Electronic address: clinicalguidelines@esmo.org. Soft tissue and visceral sarcomas: ESMO-EURACAN-GENTURIS Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021; 32(11): 1348–1365.
  135. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology, Soft Tissue Sarcoma. Wersja 2.2022. www.nccn.org (30.08.2022).
  136. Grimer R, Judson I, Peake D, et al. Guidelines for the management of soft tissue sarcomas. Sarcoma. 2010; 2010: 506182.
  137. Ruka W, Rutkowski P, Krzakowski M, et al. Mięsaki tkanek miękkich u dorosłych– zasady postępowania diagnostyczno-terapeutycznego. Nowotwory J Oncol. 2010; 60: 55–65.
  138. Sakata K, Beitler AL, Gibbs JF, et al. How surgeon age affects surveillance strategies for extremity soft tissue sarcoma patients after potentially curative treatment. J Surg Res. 2002; 108(2): 227–234.
  139. Chou YS, Liu CY, Chen WM, et al. Follow-up after primary treatment of soft tissue sarcoma of extremities: impact of frequency of follow-up imaging on disease-specific survival. J Surg Oncol. 2012; 106(2): 155–161.
  140. Geel Av, Pastorino U, Jauch K, et al. Surgical treatment of lung metastases: The European Organization for Research and Treatment of Cancer-Soft Tissue and Bone Sarcoma Group study of 255 patients. Cancer. 1996; 77(4): 675–682, doi: 10.1002/(sici)1097-0142(19960215)77:4<675::aid-cncr13>3.0.co;2-y.
  141. Patel SR, Zagars GK, Pisters PWT. The follow-up of adult soft-tissue sarcomas. Semin Oncol. 2003; 30(3): 413–416.
  142. Casson A, Putnam J, Natarajan G, et al. Five-year survival after pulmonary metastasectomy for adult soft tissue sarcoma. Cancer. 1992; 69(3): 662–668, doi: 10.1002/1097-0142(19920201)69:3<662::aid-cncr2820690311>3.0.co;2-i.
  143. Gadd MA, Casper ES, Woodruff JM, et al. Development and treatment of pulmonary metastases in adult patients with extremity soft tissue sarcoma. Ann Surg. 1993; 218(6): 705–712.
  144. Whooley BP, Gibbs JF, Mooney MM, et al. Primary extremity sarcoma: what is the appropriate follow-up? Ann Surg Oncol. 2000; 7(1): 9–14.
  145. Whooley BP, Mooney MM, Gibbs JF, et al. Effective follow-up strategies in soft tissue sarcoma. Semin Surg Oncol. 1999; 17(1): 83–87, doi: 10.1002/(sici)1098-2388(199907/08)17:1<83::aid-ssu11>3.0.co;2-w.
  146. Puri A, Gulia A, Hawaldar R, et al. Does intensity of surveillance affect survival after surgery for sarcomas? Results of a randomized noninferiority trial. Clin Orthop Relat Res. 2014; 472(5): 1568–1575.
  147. Choi H, Varma DG, Fornage BD, et al. Soft-tissue sarcoma: MR imaging vs sonography for detection of local recurrence after surgery. AJR Am J Roentgenol. 1991; 157(2): 353–358.
  148. Arya S, Nagarkatti DG, Dudhat SB, et al. Soft tissue sarcomas: ultrasonographic evaluation of local recurrences. Clin Radiol. 2000; 55(3): 193–197.
  149. Briccoli A, Galletti S, Salone M, et al. Ultrasonography is superior to computed tomography and magnetic resonance imaging in determining superficial resection margins of malignant chest wall tumors. J Ultrasound Med. 2007; 26(2): 157–162.
  150. Labarre D, Aziza R, Filleron T, et al. Detection of local recurrences of limb soft tissue sarcomas: is magnetic resonance imaging (MRI) relevant? Eur J Radiol. 2009; 72(1): 50–53.
  151. Vanel D, Shapeero LG, De Baere T, et al. MR imaging in the follow-up of malignant and aggressive soft-tissue tumors: results of 511 examinations. Radiology. 1994; 190(1): 263–268.
  152. Brennan MF. Follow-up is valuable and effective: true, true and unrelated? Ann Surg Oncol. 2000; 7(1): 2–3.
  153. Casali PG, Blay JY, Abecassis N, et al. Gastrointestinal stromal tumours: ESMO–EURACAN–GENTURIS Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2022; 33(1): 20–33.
  154. Rutkowski P, Ługowska I, Fijuth J, et al. Soft tissue sarcomas in adults. Oncol Clin Pract. 2017; 13: 181–201.
  155. Rutkowski P, Wozniak A, Dębiec-Rychter M, et al. Clinical utility of the new American Joint Committee on Cancer staging system for gastrointestinal stromal tumors: current overall survival after primary tumor resection. Cancer. 2011; 117(21): 4916–4924.
  156. Joensuu H, Eriksson M, Sundby Hall K, et al. One vs three years of adjuvant imatinib for operable gastrointestinal stromal tumor: a randomized trial. JAMA. 2012; 307(12): 1265–1272.
  157. Rutkowski P, Świtaj T, Mazurkiewicz T, et al. Bone sarcomas. Oncol Clin Pract. 2018; 14: 115–128.
  158. Strauss SJ, Frezza AM, Abecassis N, et al. ESMO Guidelines Committee, EURACAN, GENTURIS and ERN PaedCan. Electronic address: clinicalguidelines@esmo.org. Bone sarcomas: ESMO-EURACAN-GENTURIS-ERN PaedCan Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol. 2021; 32(12): 1520–1536.
  159. Lin PP, Patel S. Bone sarcoma. Springer, New York 2013.
  160. Grimer R, Athanasou N, Gerrand C, et al. UK Guidelines for the Management of Bone Sarcomas. Sarcoma. 2010; 2010: 317462.
  161. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Bone Cancer. Wersja 1.2023. www.nccn.org (30.08.2022).
  162. Bassal M, Mertens AC, Taylor L, et al. Risk of selected subsequent carcinomas in survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J Clin Oncol. 2006; 24(3): 476–483.
  163. Rodriguez-Galindo C, Poquette CA, Marina NM, et al. Hematologic abnormalities and acute myeloid leukemia in children and adolescents administered intensified chemotherapy for the Ewing sarcoma family of tumors. J Pediatr Hematol Oncol. 2000; 22(4): 321–329.
  164. Aksnes LH, Bauer H, Dahl AA, et al. Health status at long-term follow-up in patients treated for extremity localized Ewing Sarcoma or osteosarcoma: A Scandinavian sarcoma group study. Pediatr Blood Cancer. 2009; 53(1): 84–89.
  165. Langer T, Stöhr W, Paulides M, et al. Prospective multicenter registration of major late sequelae in sarcoma patients using the Late Effects Surveillance System (LESS). Klin Padiatr. 2005; 217(3): 176–181.
  166. Goryń T, Szostakowski B, Pieńkowski A, et al. Long-term follow-up in adults with extremity osteosarcoma: comparison of different surgical procedures - single-center experience. Contemp Oncol (Pozn). 2019; 23(4): 234–238.
  167. Hewitt M, Greenfield S, Stovall E. From Cancer Patient to Cancer Survivor. National Academies Press, Washington 2005.
  168. Lafata J, Simpkins J, Schultz L, et al. Routine Surveillance Care After Cancer Treatment With Curative Intent. Med Care. 2005; 43(6): 592–599.
  169. McCabe MS, Partridge AH, Grunfeld E, et al. Risk-based health care, the cancer survivor, the oncologist, and the primary care physician. Semin Oncol. 2013; 40(6): 804–812.
  170. Howell D, Hack TF, Oliver TK, et al. Models of care for post-treatment follow-up of adult cancer survivors: a systematic review and quality appraisal of the evidence. J Cancer Surviv. 2012; 6(4): 359–371.
  171. Radecka B, Streb J. Czy kontrolę po leczeniu onkologicznym powinien prowadzić onkolog? Nowotwory. 2016; 1: 274–277.
  172. Halpern MT, Viswanathan M, Evans TS, et al. Models of Cancer Survivorship Care: Overview and Summary of Current Evidence. J Oncol Pract. 2015; 11(1): e19–e27.
  173. Del Gi, Bondy SJ, Maarten S. Physician care of cancer patients: ICES atlas. Ontario 2006: 162–174.
  174. McWhinney IR, Hoddinott SN, Bass MJ, et al. Role of the family physician in the care of cancer patients. Can Fam Physician. 1990; 36: 2183–2186.
  175. Dood RL, Zhao Y, Armbruster SD, et al. Defining Survivorship Trajectories Across Patients With Solid Tumors: An Evidence-Based Approach. JAMA Oncol. 2018; 4(11): 1519–1526.
  176. Chang HM, Moudgil R, Scarabelli T, et al. Cardiovascular Complications of Cancer Therapy: Best Practices in Diagnosis, Prevention, and Management: Part 1. J Am Coll Cardiol. 2017; 70(20): 2536–2551.
  177. Hewitt M, Greenfield S, Stovall E. From Cancer Patient to Cancer Survivor: Lost in Transition. The National Academies Press, Washington 2006.
  178. Grunfeld E, Earle CC. The interface between primary and oncology specialty care: treatment through survivorship. J Natl Cancer Inst Monogr. 2010; 2010(40): 25–30.
  179. Krajowy Rejestr Nowotworów. http://onkologia.org.pl/ (30.08.2022).
  180. Nekhlyudov L, Aziz NM, Lerro C, et al. Oncologists' and primary care physicians' awareness of late and long-term effects of chemotherapy: implications for care of the growing population of survivors. J Oncol Pract. 2014; 10(2): e29–e36.
  181. Snyder CF, Earle CC, Herbert RJ, et al. Preventive care for colorectal cancer survivors: a 5-year longitudinal study. J Clin Oncol. 2008; 26(7): 1073–1079.
  182. Grunfeld E, Mant D, Vessey MP, et al. Specialist and general practice views on routine follow-up of breast cancer patients in general practice. Fam Pract. 1995; 12(1): 60–65.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Wydawcą serwisu jest VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail: viamedica@viamedica.pl