Anemias in Oncology: Potential of Maintenance Therapy

AV Snegovoi, VB Larionova, LV Manzyuk, IB Kononenko

N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Anton Vladimirovich Snegovoi, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-41-09; e-mail: anvs2012@gmail.com

For citation: Snegovoi AV, Larionova VB, Manzyuk LV, Kononenko IB. Anemias in Oncology: Potential of Maintenance Therapy. Clinical oncohematology. 2016;9(3):326-35 (In Russ).

DOI: 10.21320/2500-2139-2016-9-3-326-35


ABSTRACT

Development of anemia during chemotherapy or chemoradiotherapy of malignancies is a serious adverse event negatively affecting the quality of life and effectiveness of the treatment. In this regard, the leading consensus committees of NCCN, ESMO, ASCO, and RUSSCO have prepared and are regularly updating recommendations for diagnosing and treatment of anemia in cancer patients. The article presents recent data on the pathogenesis and methods of treatment of anemia in cancer patients, including administration of erythropoiesis stimulating agents: recombinant erythropoietins, intravenous iron products, vitamins, and red blood cell transfusions.


Keywords: cancer, anemia, erythropoietins, intravenous iron products, red blood cell transfusions.

Received: February 29, 2016

Accepted: March 31, 2016

Read in PDF(RUS)pdficon


REFERENCES

  1. Knight K, Wade S, Balducci L. Prevalence and outcome of anemia in cancer: a systematic review of the literature. Am J Med. 2004;116(7);11–26. doi: 10.1016/j.amjmed.2003.12.008.
  2. Schrijvers D, De Samblanx H, Roila F. Erythropoiesis-stimulating agents in the treatment of anaemia in cancer patients: ESMO Clinical Practice Guidelines for use. Ann Oncol. 2010;21(Suppl 5):v244–v247. doi: 10.1093/annonc/mdq202.
  3. Ludwig H, Van Belle S, Barrett-Lee P, et al. The European Cancer Anaemia Survey (ECAS): A large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur J Cancer. 2004;40(15):2293–306. doi: 10.1016/j.ejca.2004.06.019.
  4. Groopman J, Itri L. Chemotherapy-induced anemia in adults: incidence and treatment. J Nat Cancer Inst. 1999;91(19):1616–34. doi: 10.1093/jnci/91.19.1616.
  5. Food and Drug Administration. Jevtana (cabazitaxel) Injection Label Information. [Internet] Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/201023lbl.pdf. (accessed 12.11.2015).
  6. Food and Drug Administration. Taxotere (docetaxel) Injection Label Information. [Internet] Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/020449s059lbl.pdf (accessed 12.11.2015).
  7. Food and Drug Administration. Xtandi (enzalutamide) Capsules Label Information. [Internet] Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203415lbl.pdf. (accessed 12.11.2015).
  8. Cella D. Quality of life and clinical decisions in chemotherapy-induced anemia. Oncology (Williston Park). 2006;20(8 Suppl 6):25–8.
  9. Caro JJ, Salas M, Ward A, et al. Anemia as an independent prognostic factor for survival in patients with cancer: a systemic, quantitative review. Cancer. 2001;91(12):2214–21. doi: 10.1002/1097-0142(20010615)91:12<2214::aid-cncr1251>3.0.co;2-p.
  10. Glaspy J. Update on Safety of ESAs in Cancer-Induced Anemia. J Natl Compr Cancer Network. 2012;10(5):659–66.
  11. Nowrousian M, Smyth JF, et al, eds. rhErythropoietin in cancer supportive treatment. New York: Marcel Dekker Inc; 1996. pp. 13–34.
  12. Павлов А.Д., Морщакова Е.Ф., Румянцев А.Г. Эритропоэз, эритропоэтин, железо. Молекулярные и клинические аспекты. М.: ГЭОТАР-Медиа, 2011. 304 с.
    [Pavlov AD, Morshchakova EF, Rumyantsev AG. Eritropoez, eritropoetin, zhelezo. Molekulyarnye i klinicheskie aspekty. (Erythropoiesis, erythropoietin, and iron. Molecular and clinical aspects.) Moscow: GEOTAR-Media Publ.; 2011. 304 p. (In Russ)]
  13. Grotto HZ. Anaemia of cancer: an overview of mechanisms involved in its pathogenesis. Med Oncol. 2008;25(1):12–21. doi: 10.1007/s12032-007-9000-8.
  14. Aapro M, Osterborg A, Gascon P, et al. Prevalence and management of cancer-related anaemia, iron deficiency and the specific role of i.v. iron. Ann Oncol. 2012;23(8):1954–62. doi: 10.1093/annonc/mds112.
  15. Khorana AA, Francis CW, Blumberg N, et al. Blood transfusions, thrombosis and mortality in hospitalized cancer patients. Arch Int Med. 2008;168(21):2377–81. doi: 10.1001/archinte.168.21.2377.
  16. Amato A, Pescatori M. Perioperative blood transfusions for the recurrence of colorectal cancer. Cochrane database syst.rev. 2006;25(1):CD005033. doi: 10.1002/14651858.cd005033.pub2.
  17. Hoff CM, Lassen P, Eriksen JG, et al. Does transfusion improve the outcome for HNSCC patients treated with radiotherapy? Results from the randomized DAHANCA 5 and 7 trials. Acta Oncologica. 2011;50(7):1006–14. doi: 10.3109/0284186X.2011.592650.
  18. Egrie JC, Browne JK. Development and characterization of novel erythropoiesis stimulating protein (NESP). Br J Cancer. 2001;84(Suppl 1):3–10. doi: 10.1054/bjoc.2001.1746.
  19. Bohlius J, Wilson J, Seidenfeld J, et al. Recombinant human erythropoietins and cancer patients: updated meta-analysis of 57 studies including 9353 patients. J Natl Cancer Inst. 2006;98(10):708–14. doi: 10.1093/jnci/djj189.
  20. Tonelli M, Hemmelgarn B, Reiman T, et al. Benefits and harms of erythropoiesis-stimulating agents for anemia related to cancer: a meta-analysis. CMAJ. 2009;180(11):E62–E71. doi: 10.1503/cmaj.090470.
  21. Bennett C, Silver S, Djulbegovic B, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA. 2008;299(8):914–24. doi: 10.1001/jama.299.8.914.
  22. Ludwig H, Crawford J, Osterborg A, et al. Pooled analysis of individual patient-level data from all randomized, double-blind, placebo-controlled trials of darbepoetin alfa in the treatment of patients with chemotherapy-induced anemia. J Clin Oncol. 2009;27(17):2838–47. doi: 10.1200/jco.2008.19.1130.
  23. Glaspy J, Crawford J, Vansteenkiste J, et al. Erythropoiesis-stimulating agents in oncology: a study-level meta-analysis of survival and other safety outcomes. Br J Cancer. 2010;102(2):301–15. doi: 10.1038/sj.bjc.6605498.
  24. Oncologic Drug Advisory Committee (ODAC) Meeting Information Package. Darbepoetin alfa (BLA #103951) and Epoetin alfa (BLA #103234). [Internet] Available from: http://www.scribd.com/doc/1117102/US-Food-and-Drug-Administration-20074301b20101Amgen. (accessed 20.02.2016).
  25. Glaspy J, Osterborg A, Ludwig H, et al. Evaluation of the association between (Hb) events and safety outcomes in cancer patients with chemotherapy induced anemia: an integrated analysis of patient-level data from 6 randomized, placebo-controlled trials of darbepoetin. Eur J Cancer. 2007;5(4):147–8. doi: 10.1016/s1359-6349(07)70639-0.
  26. Снеговой А.В., Aapro M., Давиденко И.С. и др. Практические рекомендации по лечению анемии у онкологических больных. Злокачественные опухоли. 2015;4:316–26. doi: 10.18027/2224-5057-2015-4s-316-326. [Snegovoi AV, Aapro M, Davidenko IS, et al. Practical recommendations for management of anemia in cancer patients. Zlokachestvennye opukholi. 2015;4:316–26. doi: 10.18027/2224-5057-2015-4s-316-326. (In Russ)]
  27. Henke M, Laszig R, Rube C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. The Lancet. 2003;362(9392):1255–60. doi: 10.1016/s0140-6736(03)14567-9.
  28. Leyland-Jones B, Semiglazov V, Pawlicki M, et al. Maintaining normal hemoglobin levels with epoetin alfa in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. J Clin Oncol. 2005;23(25):5960–72. doi: 10.1200/jco.2005.06.150.
  29. Delarue R, Haioun C, Coiffier B, et al. Survival effect of darbepoetin alfa in patients with diffuse large B-cell lymphoma (DLBCL) treated with immunochemotherapy: The LNH03-6B study. J Clin Oncol. 2011;29: Abstract 9048.
  30. Nitz U, Gluz O, Oberhoff C, et al. Adjuvant chemotherapy with or without darbepoetin alpha in node-positive breast cancer: survival and quality of life analysis from the prospective randomized WSG ARA plus trial. Cancer Res. 2012;71(24 Suppl):PD07-06. doi: 10.1158/0008-5472.sabcs11-pd07-06.
  31. National Institute for Health and Care Excellence. Health Technology Appraisal Programme. Equality impact assessment – Guidance development. MTA Erythropoiesis-stimulating agents (epoetin and darbepoetin) for treating anaemia in people having cancer treatment (including review of TA142). [Internet] Available from: https://www.nice.org.uk/guidance/ta323/documents/anaemia-cancertreatment-induced-erythropoiesisstimulating-agents-epoetin-and-darbepoetin-inc-rev-ta142-equality-impact-assessment-guidance-development2. (accessed 06.06.2016).
  32. Aapro M, Osterborg A, Gascon P, et al. Prevalence and management of cancer-related anaemia, iron deficiency and the specific role of i.v. iron. Ann Oncol. 2012;23(8):1954–62. doi: 10.1093/annonc/mds112.
  33. Beale AL, Penney MD, Allison MC. The prevalence of iron deficiency among patients presenting with colorectal cancer. Colorectal Dis. 2005;7(4):398–402. doi: 10.1111/j.1463-1318.2005.00789.x.
  34. Kuvibidila S, Gauthier T, Rayford W. Increased levels of serum transferrin receptor and serum transferrin receptor/log ferritin ratios in men with prostate cancer and the implications for body-iron stores. J Lab Clin Med. 2004;144(4):176–82. doi: 10.1016/j.lab.2004.03.017.
  35. Steinmetz HT, Tsamaloukas A, Schmitz S, et al. A new concept for the differential diagnosis and therapy of anaemia in cancer patients. Supp Care Cancer. 2010;19(2):261–9. doi: 10.1007/s00520-010-0812-2.
  36. Beguin Y, Lybaert W, Bosly A. A prospective observational study exploring the impact of iron status on response to darbepoetin alfa in patients with chemotherapy induced anemia. Blood. 2009;114(22): Abstract 2007.
  37. Ludwig H, Muldur E, Endler G, et al. High prevalence of iron deficiency across different tumors correlates with anemia, increases during cancer treatment and is associated with poor performance status. Haematologica. 2011;96: Abstract 982.
  38. Dangsuwan P, Manchana T. Blood transfusion reduction with intravenous iron in gynecologic cancer patients receiving chemotherapy. Gynecol Oncol. 2010;116(3):522–5. doi: 10.1016/j.ygyno.2009.12.004.
  39. Kim YT, Kim SW, Yoon BS, et al. Effect of intravenously administered iron sucrose on the prevention of anemia in the cervical cancer patients treated with concurrent chemoradiotherapy. Gynecol Oncol. 2007;105(1):199–204. doi: 10.1016/j.ygyno.2006.11.014.
  40. Steinmetz HT. The role of intravenous iron in the treatment of anemia in cancer patients. Ther Adv Hematol. 2012;3(3):177–91. doi: 10.1177/ 2040620712440071.
  41. Henry DH. Parenteral Iron Therapy in Cancer-Associated Anemia. Hematology. 2010;2010(1):351–6. doi: 10.1182/asheducation-2010.1.351.
  42. Auerbach M, Silberstein PT, Webb RT, et al. Darbepoetin alfa 300 or 500 mg once every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. Am J Hematol. 2010;85(9):655–63. doi: 10.1002/ajh.21779.
  43. Bastit L, Vandebroek A, Altintas S, et al. Randomized, multicenter, controlled trial comparing the efficacy and safety of darbepoetin alpha administered every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. J Clin Oncol. 2008;26(10):1611–8. doi: 10.1200/jco.2006.10.4620.
  44. Henry DH, Dahl NV, Auerbach M, et al. Intravenous ferric gluconate significantly improves response to epoetin alfa versus oral iron or no iron in anemic patients with cancer receiving chemotherapy. The Oncologist. 2007;12(2):231–42. doi: 10.1634/theoncologist.12-2-231.
  45. Pedrazzoli P, Farris A, Del Prete S, et al. Randomized trial of intravenous iron supplementation in patients with chemotherapy-related anemia without iron deficiency treated with darbepoetin alpha. J Clin Oncol. 2008;26(10):1619–25. doi: 10.1200/jco.2007.12.2051.
  46. Steensma DP, Sloan JA, Dakhil SR, et al. Phase III, randomized study of the effects of parenteral iron, oral iron, or no iron supplementation on the erythropoietic response to darbepoetin alfa for patients with chemotherapy-associated anemia. J Clin Oncol. 2011;29(1):97–105. doi: 10.1200/jco.2010.30.3644.
  47. Beguin Y, Maertens J, De Prijck B, et al. Darbepoetin-alfa and I.V. iron administration after autologous hematopoietic stem cell transplantation: a prospective randomized multicenter trial. Am J Hematol. 2013;88(12):990–6. doi: 10.1002/ajh.23552.
  48. Hedenus M, Birgegard G, Nasman P, et al. Addition of intravenous iron to epoetin beta increases hemoglobin response and decreases epoetin dose requirement in anemic patients with lymphoproliferative malignancies: a randomized multicenter study. Leukemia. 2007;21:627–32. doi: 10.1038/sj.leu.2404562.
  49. Gafter-Gvili A, Rozen-Zvi B, Vidal L, et al. Intravenous iron supplementation for the treatment of cancer-related anemia—systematic review and meta-analysis. Acta Oncologica. 2013;52(1):18–29. doi: 10.3109/0284186x.2012.702921.
  50. Petrelli F, Borgonovo K, Cabiddu M, et al. Addition of iron to erythropoiesis-stimulating agents in cancer patients: a meta-analysis of randomized trials. J Cancer Res Clin Oncol. 2012;138(2):179–87. doi: 10.1007/s00432-011-1072-3.
  51. Сельчук В.Ю., Чистяков С.С., Толокнов Б.О. и др. Железодефицитная анемия: современное состояние проблемы. Русский медицинский журнал. 2012;3(1):1–8.
    [Sel’chuk VYu, Chistyakov SS, Toloknov BO, et al. Iron-deficiency anemia: state-of-the-art of the problem. Russkii meditsinskii zhurnal. 2012;3(1):1–8. (In Russ)]

Prevention of Nausea and Vomiting in Oncology

AV Snegovoi, VB Larionova, IB Kononenko, LV Manzyuk, AI Saltanov, VYu Sel’chuk

N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Anton Vladimirovich Snegovoi, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-41-09; e-mail: anvs2012@gmail.com

For citation: Snegovoi AV, Larionova VB, Kononenko IB, et al. Prevention of Nausea and Vomiting in Oncology. Clinical oncohematology. 2016;9(1):75–83 (In Russ).

DOI: 10.21320/2500-2139-2016-9-1-75-83


ABSTRACT

Background & Aims. Despite significant progress achieved over last 25 years, nausea and especially vomiting remain basic side effects of chemotherapy affecting patients’ quality of life. Modern antiemetic therapy permits to reduce the frequency of acute vomiting to 20–30 %, whereas the effectiveness of nausea prevention is higher. Available domestic publications do not describe fundamentals of the antiemetic therapy during treatment with cytostatic agents. The objective of this study is to evaluate the effectiveness of itopride in combined antiemetic therapy for prevention of acute nausea and vomiting, as well as in single drug therapy for arresting delayed vomiting.

Methods. 30 patients were enrolled in the study (breast cancer — 10 patients, ovarian cancer — 8 patients, gastric cancer — 2 patients, colorectal cancer — 5 patients, and lung cancer — 5 patients): 25 women and 5 men aged from 40 to 70 years. All patients received chemotherapy earlier, which had unfavorable effects on the tolerability of subsequent therapy. All patients underwent chemotherapy with different degrees of emetogenicity during the study.

Results. The study has demonstrated that overall effectiveness of a combination of itopride with 5-HT3 receptor antagonists for prevention of acute vomiting was equal to 100 %. Overall effectiveness of itopride for prevention of delayed vomiting was equal to 80 %.

Conclusion. The combination of itopride with 5-HT3 receptor antagonists can be recommended for prevention of delayed vomiting in moderately emetogenic chemotherapeutic regimens.


Keywords: nausea, vomiting, oncology, antiemetics.

Received: August 10, 2015

Accepted: October 12, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Scotte F. The Importance of Supportive Care in Optimizing Treatment Outcomes of Patients with Advanced Prostate Cancer. The Oncologist. 2012;17(Suppl. 1):23–30. doi: 10.1634/theoncologist.2012-s1-23.
  2. Warr D. Chemotherapy- and cancer-related nausea and vomiting. Curr Oncol. 2008;15(Suppl. 1):S4–9. doi: 10.3747/co.2008.171.
  3. Rolia F, Herrstedt J, Aapro M, et al. Guideline update for MASCC and ESMO in the prevention of chemotherapy- and radiotherapy-induced nausea and vomiting: results of the Perugia consensus conference. Ann Oncol. 2010;5:v232–43. doi: 10.1093/annonc/mdq194.
  4. Rojas C, Slusher BS. Pharmacological mechanisms of 5-HT3 and tachykinin NK1 receptor antagonism to prevent chemotherapy-induced nausea and vomiting. Eur J Pharmacol. 2012;684:1–7. doi: 10.1016/j.ejphar.2012.01.046.
  5. Herrstedt J. Antiemetics: an update and the MASCC guidelines applied in clinical practice. J Nat Clin Pract Oncol. 2008;5(1):32–43. doi: 10.1038/ncponc1021.
  6. Pater JL, Lofters WS, Zee B, et al. The role of the 5-HT3 antagonists ondansetron and dolasetron in the control of delayed onset nausea and vomiting in patients receiving moderately emetogenic chemotherapy. Ann Oncol. 1997;8:181–5.
  7. The Italian Group for Antiemetic Research. Dexamethasone, granisetron, or both for the prevention of nausea and vomiting during chemotherapy for cancer. N Engl J Med. 1995;332:1–5. doi: 10.1056/nejm199501053320101.
  8. Carmichael J, Bessell EM, Harris AL, et al. Comparison of granisetron alone and granisetron plus dexamethasone in the prophylaxis of cytotoxic-induced emesis. Br J Cancer. 1994;70:1161–4. doi: 10.1038/bjc.1994.465.
  9. Ioannidis JP, Hesketh PJ, Lau J. Contribution of dexamethasone to control of chemotherapy-induced nausea and vomiting: a meta-analysis of randomized evidence. J Clin Oncol. 2000;18(19):3409–22.
  10. Geling O, Eichler HG. Should 5-Hydroxytryptamine-3 Receptor Antagonists Be Administered Beyond 24 Hours After Chemotherapy to Prevent Delayed Emesis? Systematic Re-Evaluation of Clinical Evidence and Drug Cost Implications. J Clin Oncol. 2005;23(6):1289–94. doi: 10.1200/jco.2005.04.022.
  11. Italian Group for Antiemetic Research. Dexamethasone alone or in combination with ondansetron for the prevention of delayed nausea and vomiting induced by chemotherapy. N Engl J Med. 2000;342:1554–9. doi: 10.1056/nejm200005253422102.
  12. Jantunen IT, Kataja VV, Muhonen TT. An overview of randomised studies comparing 5-HT3 receptor antagonists to conventional anti-emetics in the prophylaxis of acute chemotherapy-induced vomiting. Eur J Cancer. 1997;33(1):66–74. doi: 10.1016/s0959-8049(96)00276-6.
  13. De Wit R, de Boer AC, van Linden GHM, et al. Effective cross-over to granisetron after failure to ondansetron, a randomized double-blind study in patients failing ondansetron plus dexamethasone during the first 24 hours following highly emetogenic chemotherapy. Br J Cancer. 2001;85:1099–101.
  14. Passik SD, Loehrer PJ, Navari RJ, et al. A phase I trial of olanzapine (Zyprexa) for the prevention of delayed emesis in cancer patients receiving chemotherapy: A Hoosier Oncology Group study. Proc ASCO. 2002;21:374. doi: 10.1081/cnv-200029066.
  15. Aapro MS, Thuerlimann B, Sessa C, et al. A randomized double-blind trial to compare the clinical efficacy of granisetron with metoclopramide, both combined with dexamethasone in the prophylaxis of chemotherapy-induced delayed emesis. Ann Oncol. 2003;14:291–7. doi: 10.1093/annonc/mdg075.
  16. Roila F, Donati D, Tamberi S, Margutti G. Delayed emesis: incidence, pattern, prognostic factors and optimal treatment. Supp Care Cancer. 2002;10:88–95. doi: 10.1007/s005200100295.
  17. Grunberg SM, Osoba D, Hesketh PJ, et al. Evaluation of new antiemetic agents and definition of antineoplastic agent emetogenicity – an update. Supp Care Cancer. 2005;13:80–4. doi: 10.1007/s00520-004-0718-y.
  18. Eisenberg P, MacKintosh FR, Ritch P, Cornett PA, Macciocchi A. Efficacy, safety and pharmacokinetics of palonosetron in patients receiving highly emetogeric, cisplatin-based chemotherapy: a dose-ranging, clinical study. Ann Oncol. 2004;15:330–7. doi: 10.1093/annonc/mdh047.
  19. Rubenstein EB, Slusher BS, Rojas C, Navari RM. New approaches to chemotherapy-induced nausea and vomiting: from neuropharmacology to clinical investigations. Cancer J. 2006;12:341–7. doi: 10.1097/00130404-200609000-00003.
  20. Saito M, Aogi K, Sekine I, et al. Palonosetron plus dexamethasone versus granisetron plus dexamethasone for prevention of nausea and vomiting during chemotherapy: a double-blind, double-dummy, randomised, comparative phase III trial. Lancet Oncol. 2009;10:115–24. doi: 10.1016/s1470-2045(08)70313-9.
  21. Likhun Z, Xiang J, Xin D, Tao ZL. A Systematic Review and Meta-Analysis of Intravenous Palonosetron in the Prevention of Chemotherapy-Induced Nausea and Vomiting in Adults. The Oncologist. 2011;16:207–16. doi: 10.1634/theoncologist.2010-0198.
  22. Navari RM, Nagy CK, Gray SE. The use of olanzapine versus metoclopramide for the treatment of breakthrough chemotherapy-induced nausea and vomiting in patients receiving highly emetogenic chemotherapy. Supp Care Cancer. 2013;21(6):1655–63. doi: 10.1007/s00520-012-1710-6.
  23. Herrstedt J, Aapro MS, Smyth JF, Del Favero A. Corticosteroids, dopamine antagonists and other drugs. Supp Care Cancer. 1998;6:204–14. doi: 10.1007/s005200050155.
  24. Tremblay PB, Kaiser R, Sezer O, et al. Variations in the 5-Hydroxytryptamine Type 3B Receptor Gene as Predictors of the Efficacy of Antiemetic Treatment in Cancer Patients. J Clin Oncol. 2003;21(11):2147–55. doi: 10.1200/jco.2003.05.164.
  25. Kaiser R, Tremblay PB, Sezer O, et al. Investigation of the association between 5-HT3A receptor gene polymorphisms and efficiency of antiemetic treatment with 5-HT3 receptor antagonists. Pharmacogenetics. 2004;14(5):271–8. doi: 10.1097/00008571-200405000-00001.
  26. Emend: EPAR. Scientific Discussion. European Medicine Agency; 2004. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Scientific_Discussion/human/000527/WC500026534.pdf.
  27. Grunberg S, Chua D, Maru A, et al. Single-Dose Fosaprepitant for the Prevention of Chemotherapy-Induced Nausea and Vomiting Associated With Cisplatin Therapy: Randomized, Double-Blind Study Protocol – EASE. J Clin Oncol. 2011;29(11):1495. doi: 10.1200/jco.2010.31.7859.
  28. Ando Y, Hayashi T, Ito K, et al. Comparison between 5-day aprepitant and single-dose fosaprepitant meglumine for preventing nausea and vomiting induced by cisplatin-based chemotherapy. Supp Care Cancer. 2016; 24(2): 871–8. doi: 10.1007/s00520-015-2856-9.
  29. Hesketh P, Rossi G, Rizzi G, et al. Efficacy and safety of NEPA, an oral combination of netupitant and palonosetron, for prevention of chemotherapy-induced nausea and vomiting following highly emetogenic chemotherapy: a randomized dose-ranging pivotal study. Ann Oncol. 2014;25:1340–6. doi: 10.1093/annonc/mdu110.
  30. Gralla R, Bosnjak S, Hontsa A, et al. A phase 3 study evaluating the safety and efficacy of NEPA, a fixed-dose combination of netupitant and palonosetron, for prevention of chemotherapy-induced nausea and vomiting over repeated cycles of chemotherapy. Ann Oncol. 2014;25(7):1333–9. doi: 10.1093/annonc/mdu096.
  31. Снеговой А.В., Манзюк Л.В. Роль ганатона в антиэметической терапии: новые возможности. Фарматека. 2011;20:85–8.
    [Snegovoi AV, Manzyuk LV. Role of ganaton in antiemetic therapy: new opportunities. Farmateka. 2011;20:85–8. (In Russ)]
  32. Снеговой А.В., Абрамов М.Е., Булавина И.С. и др. Практические рекомендации по профилактике и лечению тошноты и рвоты у онкологических больных. Злокачественные опухоли. 2015;4(спецвыпуск):327–37. doi: 10.18027/2224-5057-2015-4s-327-337.
    [Snegovoi AV, Abramov ME, Bulavina IS, et al. Clinical guidelines for preventing nausea and vomiting in oncological patients. Zlokachestvennie opuholi. 2015;4(special issue):327–37. doi: 10.18027/2224-5057-2015-4s-327-337. (in Russ)]

Peripherally Inserted Central Catheters: Own Experience of Clinical Use

A.M. Pronina, I.A. Kurmukov, O.A. Obuhova, Sh.R. Kashiya

N.N. Blokhin Russian Cancer Research Center under the Ministry of Health of the Russian Federation, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Anna Mikhailovna Pronina, MD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-62-59; e-mail: belmar9@yandex.ru

For citation: Pronina AM, Kurmukov IA, Obukhova OA, Kashiya ShR. Peripherally Inserted Central Catheters: Own Experience of Clinical Use. Clinical oncohematology. 2015;8(4):428–435 (In Russ).

DOI: 10.21320/2500-2139-2015-8-4-428-435


ABSTRACT

Background & Aims. At present, peripherally inserted central catheters (PICC) are widely used as a medium-term vascular access for patients with various diseases. In this country, these catheters are used rarely to provide vascular access in patients with cancer. Therefore, the purpose of the study is to formulate indications for implantation of these catheters to oncological patients taking into account our own clinical experience in implantation and operation of PICC.

Methods. From November, 2013, till November, 2014, at N.N. Blokhin Russian Cancer Research Center, 19 peripherally inserted central catheters were implanted to 19 patients. All catheters were placed by anesthesiologists. Peripheral or deep veins of a shoulder were punctured under the dynamic ultrasonic control.

Results. Successful catheterization was achieved in 18 of 19 patients. The median PICC dwell time was 33 days (range from 1 to 116 days), a total of 692 catheter days. In one case, during the implantation of a catheter, some technical complication were observed which made impossible its further using. Complications related to the catheter were reported in 7 cases including 2 cases of local infection of the soft tissue at the stationary point of a catheter. Inadvertent removal of a catheter was registered in 3 cases. No complications were registered.

Conclusion. Implantation of peripheral inserted central catheters under dynamic ultrasonic control has both high percent of a successful сatheterization and a minimum number of procedure-related complications. After training the staff and patients in operation of the catheter, this venous access allows to perform all necessary courses of treatments of up to 6 months.


Keywords: venous access, peripherally inserted central catheter, ultrasound-guided venipuncture.

Received: March 25, 2015

Accepted: September 30, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Abedin S, Kapoor G. Peripherally inserted central venous catheters are a good option for prolonged venous access in children with cancer. Pediatr Blood Cancer. 2008;51:251–5.
  2. Royal College of Nursing (2005b) IV Therapy Forum. Standards for Infusion Therapy. London: Royal College of Nursing; 2005.
  3. Petersen J, Delaney JH, Brakstad MT, et al. Silicone venous access devices positioned with their tips high in the superior vena cava are more likely to malfunction. Am J Surg. 1999;178(1):38–41. doi: 10.1016/s0002-9610(99)00124-5.
  4. Bard Access Systems, Groshong PICC. Instructions for Use.
  5. National Association of Vascular Access Networks. NAVAN Position Statement. J. Vasc. Access Device. 1998;3(2):8–10.
  6. Yamamoto AJ, Solomon JA, Soulen MC, et al. Sutureless securement device reduces complications of peripherally inserted central venous catheters. J Vasc Interv Radiol. 2002;13(1); 77–81. doi: 10.1016/s1051-0443(07)60012-8.
  7. Ahn DH, Illum HB, Wang DH, et al. Upper Extremity Venous Thrombosis in Patients With Cancer With Peripherally Inserted Central Venous Catheters: A Retrospective Analysis of Risk Factors. J Oncol Pract. 2012;9(1):8–12. doi: 10.1200/jop.2012.000595.
  8. Chopra V, Ratz D, Kuhn L, et al. Peripherally inserted central catheter-related deep vein thrombosis: contemporary patterns and predictors. J Thromb Haemost. 2014;12(6):847–54. doi: 10.1111/jth.12549.
  9. Liem TK, Yanit KE, Moseley SE, et al. Peripherally inserted central catheter usage patterns and associated symptomatic upper extremity venous thrombosis. J Vasc Surg. 2012;55(3):761–7. doi: 10.1016/j.jvs.2011.10.005.
  10. Kirkpatrick A, Rathbun S, Whitsett T, Raskob G. Prevention of central venous catheter-associated thrombosis: a meta-analysis. Am J Med. 2007;120(10):901.e1–13. doi: 10.1016/j.amjmed.2007.05.010.
  11. Kuter DJ. Thrombotic complications of central venous catheters in cancer patients. The Oncologist. 2004;9:207–16. doi: 10.1634/theoncologist.9-2-207.
  12. Kenney BD, David M, Bensoussan AL. Anticoagulation without catheter removal in children with catheter-related central vein thrombosis. J Pediatr Surg. 1996;31:816–8. doi: 10.1016/s0022-3468(96)90141-4.
  13. Tran H, Arellano M, Chamsuddin A, et al. Deep venous thromboses in patients with hematological malignancies after peripherally inserted central venous catheters. Leuk Lymphoma. 2010;51:1473–7. doi: 10.3109/10428194.2010.481065.
  14. Kovacs MJ, Kahn SR, Rodger M, et al. A pilot study of central venous catheter survival in cancer patients using low-molecular-weight heparin (dalteparin) and warfarin without catheter removal for the treatment of upper extremity deep vein thrombosis. J Thromb Haemost. 2007;5:1650–3. doi: 10.1111/j.1538-7836.2007.02613.x.
  15. Maki DG, Kluger DM, Crnich CJ. The risk of bloodstream infection in adults with different intravascular devices: a systematic review of 200 published prospective studies. Mayo Clin Proc 2006;81(9):1159–71. doi: 10.4065/81.9.1159.
  16. Chopra V, O’Horo JC, Rogers MA, Maki DG, Safdar N. The risk of bloodstream infection associated with peripherally inserted central catheters compared with central venous catheters in adults: a systematic review and meta-analysis. Infect Contr Hosp Epidemiol. 2013;34(9):908–18. doi: 10.1086/671737.
  17. Vidal V, Muller C, Jacquier A, et al. Prospective evaluation of PICC line related complications. J Radiol. 2008;89(4):495–8. doi: 10.1016/s0221-0363(08)71453-7.
  18. Merrell SW, Peatross BG, Grossman MD, et al. Peripherally inserted central venous catheters. Low-risk alternatives for ongoing venous access. West J Med. 1994;160:25–30.
  19. Hoffer AK, Bloch RD, Borsa JJ, et al. Peripherally inserted central venous catheters with distal versus proximal valves: prospective randomized trial. J Vasc Interv Radiol. 2001;12:1173–7. doi: 10.1016/s1051-0443(07)61676-5.
  20. Ryder MA. Peripherally inserted central venous catheters. Nurs Clin North Am. 1993;28(4):937–71.
  21. Gallieni M, Pittiruti M, Biffi R. Vascular Access in Oncology Patients. CA Cancer J Clin. 2008;58:323–46. doi: 10.3322/ca.2008.0015.

 

Anorexia-Cachexia Syndrome in Cancer Patients

AV Snegovoi, IB Kononenko, VB Larionova, AI Saltanov, VYu Sel’chuk

N.N. Blokhin Russian Cancer Research Center, 24 Kashirskoye sh., Moscow, Russian Federation, 115478

For correspondence: Anton Vladimirovich Snegovoi, PhD, 24 Kashirskoye sh., Moscow, Russian Federation, 115478; Tel.: +7(499)324-41-09; e-mail: anvs2012@gmail.com

For citation: Snegovoi AV, Kononenko IB, Larionova VB, et al. Anorexia-Cachexia Syndrome in Cancer Patients. Clinical oncohematology. 2015;8(2):185–90 (In Russ).


ABSTRACT

For a long time, cachexia has been considered one of symptoms of cancer progression. The word cachexia is of Greek origin (kakos — bad and hexis — condition). This disorder is associated with a decrease in physical activity, reduced effectiveness of anticancer therapy, and decreased survival rates. The study of dissemination of malignancies demonstrated that cachexia is caused not only by the malignancy itself, but also by development of anorexia (loss of appetite) due to changes in humoral regulation of metabolic processes. Therefore, such condition of cancer patients should be called an anorexia-cachexia syndrome.


Keywords: anorexia-cachexia syndrome, cachexia, anorexia.

Received: February 3, 2015

Accepted: February 10, 2015

Read in PDF (RUS)pdficon


REFERENCES

  1. Кононенко И.Б., Ларионова В.Б., Манзюк Л.В. и др. Синдром анорексии-кахексии. В кн.: Практические рекомендации по лекарственному лечению злокачественных опухолей (RUSSCO). Под ред. В.М. Моисеенко. М.: Общество онкологов-химиотерапевтов, 2014. С. 399–402.
    [Kononenko IB, Larionova VB, Manzyuk LV, et al. Cahexia-anorexia syndrome. In: Moiseenko VM, ed. Prakticheskie rekomendatsii po lekarstvennomu lecheniyu zlokachestvennykh opukholei (RUSSCO). (Practical guidelines for drug therapy of malignancies (RUSSCO).) Moscow: Obshchestvo onkologov-khimioterapevtov Publ.; 2014. pp. 399–402. (In Russ)]
  2. DeVita VT. Jr, Lawrence TS, Rosenberg SA. Cancer: Principles and Practice of Oncology. 9th edition. Philadelphia: Lippincott Williams & Wilkins; 2011.
  3. Dewys WD, Begg C, Lavin PT, et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. Am J Med. 1980;69(4):491–7. doi: 10.1016/s0149-2918(05)80001-3.
  4. Del Fabbro E, Inui A, Strasser F. Cancer Cachexia. Pocket book for cancer supportive care. Springer Healthcare; 2012.
  5. Снеговой А.В. Школа «Поддерживающей терапии в онкологии». 2014. http://www.rosoncoweb.ru/events/2014/12/05/
    [Snegovoi AV. Maintenance Therapy in Oncology School; 2014. http://www.rosoncoweb.ru/events/2014/12/05/]
  6. Hamid RAH, Umbas R, Mochtar CA. Recent role of inflammation in prostate diseases: chemoprevention development opportunity. Acta Med Indones. 2011;43(1):59–65.
  7. Argiles JM, Busquets S, Garcia-Martonez C, Lopez-Soriano FJ. Mediators involved in the cancer anorexia-cachexia syndrome: past, present, and future. Nutrition. 2005;21(9):977–85. doi: 10.1016/j.nut.2005.02.003.
  8. Materials of conference of the Multinational Association of Supportive Care in Cancer (MASCC), June 2014. J Sup Care Cancer. 2014;22(Suppl 1):28–31.
  9. Fearon K, Strasser F, Anker SD, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489–95. doi: 10.1016/s1470-2045(10)70218-7.
  10. Evans WJ, Morley JE, Argiles J, et al. Cachexia: a new definition. J Clin Nutr. 2008;27(6):793–9. doi: 10.1016/j.clnu.2008.06.013.
  11. Daley RJ, Canada T. Managing the cancer anorexia-cachexia syndrome: a pharmacologic review. Oncol Nutr Connect. 2004;12(4):1–6.
  12. Bosaeus I. Nutritional support in multimodal therapy for cancer cachexia. Sup Care Cancer. 2008;16(5):447–51. doi: 10.1007/s00520-007-0388-7.
  13. Thoresen L, Frykholm G, Lydersen S, et al. Nutritional status, cachexia and survival in patients with advanced colorectal carcinoma. Different assessment criteria for nutritional status provide unequal results. Clin Nutr. 2013;32(1):65–72. doi: 10.1016/j.clnu.2012.05.009.
  14. Baldwin C, Spiro A, McGough C, et al. Simple nutritional intervention in patients with advanced cancers of the gastrointestinal tract, non-small cell lung cancers or mesothelioma and weight loss receiving chemotherapy: a randomised controlled trial. J Hum Nutr Diet. 2011;24(5):431–40. doi: 10.1111/j.1365-277x.2011.01189.x.
  15. Maltoni M, Nanni O, Scarpi E. High-dose progestins for the treatment of cancer anorexia-cachexia syndrome: A systematic review of randomised clinical trials. Ann Oncol. 2001;12(3):289–300. doi: 10.1023/a:1011156811739.