201
|
Engert A, Josting A, Haverkamp H, Villalobos M, Lohri A, Sökler M, Zijlstra J, Sturm I, Topp MS, Rank A, Zenz T, Vogelhuber M, Nogova L, Borchmann P, Fuchs M, Flechtner HH, Diehl V. Epoetin Alfa in Patients With Advanced-Stage Hodgkin's Lymphoma: Results of the Randomized Placebo-Controlled GHSG HD15EPO Trial. J Clin Oncol 2010; 28:2239-45. [DOI: 10.1200/jco.2009.25.1835] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose To determine whether epoetin alfa reduces anemia-related fatigue, improves other aspects of health-related patient-recorded outcomes (PROs), reduces the number of RBC transfusions, and has an impact on freedom from treatment failure (FFTF) and overall survival (OS) in patients with advanced-stage Hodgkin's lymphoma (HL). Patients and Methods The prospectively randomized HD15EPO study performed by the German Hodgkin Study Group investigated epoetin alfa administered at doses of 40,000 U weekly during and after chemotherapy (six to eight cycles of bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone [BEACOPP]) in a double-blind, placebo-controlled setting. The study accrued 1,379 patients, of whom 1,328 were assessable for safety, 1,303 were assessable for clinical outcome, and 930 were assessable for PROs. Results PROs were not different in patients receiving placebo or epoetin alfa, both after the end of chemotherapy and 6 months thereafter. There was no difference between patients treated with epoetin alfa or placebo with respect to FFTF and OS. There were also no differences in the numbers of deaths, progressions, relapses, and thromboembolic events. The median number of RBC transfusions was reduced from four per patient in the placebo group to two per patient in the epoetin alfa group (P < .001), with 27.4% of patients needing no RBC transfusion in the placebo group compared with 36.7% of patients in the epoetin alfa group (P < .001). Conclusion Epoetin alfa administered at 40,000 U weekly parallel to BEACOPP chemotherapy was safe in patients with advanced-stage HL and reduced the number of RBC transfusions but had no impact on fatigue and other PRO domains.
Collapse
Affiliation(s)
- Andreas Engert
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Andreas Josting
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Heinz Haverkamp
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Matthias Villalobos
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Andreas Lohri
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Martin Sökler
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Josée Zijlstra
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Isrid Sturm
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Max S. Topp
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Andreas Rank
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Thorsten Zenz
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Martin Vogelhuber
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Lucia Nogova
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Peter Borchmann
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Michael Fuchs
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Hans-Henning Flechtner
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| | - Volker Diehl
- From the First Department of Internal Medicine; German Hodgkin Study Group, University of Cologne, Cologne; Department of Internal Medicine V, University of Heidelberg, Heidelberg; Department of Hematology/Oncology, University of Tübingen, Tübingen; Department of Hematology and Oncology, Charité-Campus Virchow, Berlin; Medical Clinic and Polyclinic II, Julius-Maximilian University of Würzburg, Würzburg; Department of Internal Medicine III – Großhadern, Ludwig-Maximilians-University, Munich; Department of
| |
Collapse
|
202
|
Szenajch J, Wcislo G, Jeong JY, Szczylik C, Feldman L. The role of erythropoietin and its receptor in growth, survival and therapeutic response of human tumor cells From clinic to bench - a critical review. Biochim Biophys Acta Rev Cancer 2010; 1806:82-95. [PMID: 20406667 DOI: 10.1016/j.bbcan.2010.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 04/05/2010] [Accepted: 04/11/2010] [Indexed: 12/27/2022]
Abstract
Recombinant human erythropoietin (rhEPO) has been used clinically to alleviate cancer- and chemotherapy-related anemia. However, recent clinical trials have reported that rhEPO also may adversely impact disease progression and survival. The expression of functional EPO receptors (EPOR) has been demonstrated in many human cancer cells where, at least in vitro, rhEPO can stimulate cell growth and survival and may induce resistance to selected therapies. Responses to rhEPO measured by alterations in tumor cell growth or survival, activation of signaling pathways or modulation of sensitivity to anticancer agents are variable. Both methodological and inherent biological issues underlie the differential cell responses, including reported difficulties in EPOR protein detection, potential involvement of EPOR isoforms or of cytoplasmic EPOR, possible differential structure and/or binding affinities of hematopoietic versus non-hematopoietic cell EPOR, possible aberrant regulation of EPOR activity, and a functional EPO/EPOR autocrine/paracrine loop. The modulation by rhEPO of tumor cell response to anticancer agents is coincident with modulation of multiple signaling pathways, BCL-2 family proteins, caspases and NFkB. The molecular interplay of pro-survival and pro-death signals, triggered by EPO and/or by anticancer agents, is multifactorial and tightly coordinated. Expression microarray analysis may prove critical for deciphering this potentially novel network and its broad spectrum of genes and proteins.
Collapse
Affiliation(s)
- Jolanta Szenajch
- Laboratory for Molecular Oncology, Military Institute of Medicine, Warsaw, Poland
| | | | | | | | | |
Collapse
|
203
|
Lyman GH, Kuderer NM. Prevention and treatment of venous thromboembolism among patients with cancer: The American Society of Clinical Oncology Guidelines. Thromb Res 2010; 125 Suppl 2:S120-7. [DOI: 10.1016/s0049-3848(10)70029-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
204
|
Shi Z, Hodges VM, Dunlop EA, Percy MJ, Maxwell AP, El-Tanani M, Lappin TRJ. Erythropoietin-induced activation of the JAK2/STAT5, PI3K/Akt, and Ras/ERK pathways promotes malignant cell behavior in a modified breast cancer cell line. Mol Cancer Res 2010; 8:615-626. [PMID: 20353997 DOI: 10.1158/1541-7786.mcr-09-0264] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Erythropoietin (Epo), the major regulator of erythropoiesis, and its cognate receptor (EpoR) are also expressed in nonerythroid tissues, including tumors. Clinical studies have highlighted the potential adverse effects of erythropoiesis-stimulating agents when used to treat cancer-related anemia. We assessed the ability of EpoR to enhance tumor growth and invasiveness following Epo stimulation. A benign noninvasive rat mammary cell line, Rama 37, was used as a model system. Cell signaling and malignant cell behavior were compared between parental Rama 37 cells, which express few or no endogenous EpoRs, and a modified cell line stably transfected with human EpoR (Rama 37-28). The incubation of Rama 37-28 cells with pharmacologic levels of Epo led to the rapid and sustained increases in phosphorylation of signal transducers and activators of transcription 5, Akt, and extracellular signal-regulated kinase. The activation of these signaling pathways significantly increased invasion, migration, adhesion, and colony formation. The Epo-induced invasion capacity of Rama 37-28 cells was reduced by the small interfering RNA-mediated knockdown of EpoR mRNA levels and by inhibitors of the phosphoinositide 3-kinase/Akt and Ras/extracellular signal-regulated kinase signaling pathways with adhesion also reduced by Janus-activated kinase 2/signal transducers and activators of transcription 5 inhibition. These data show that Epo induces phenotypic changes in the behavior of breast cancer cell lines and establishes links between individual cell signaling pathways and the potential for cancer spread.
Collapse
Affiliation(s)
- Zhanzhong Shi
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
205
|
Thromboembolic events in patients with colorectal cancer receiving the combination of bevacizumab-based chemotherapy and erythropoietin stimulating agents. Am J Clin Oncol 2010; 33:36-42. [PMID: 19652579 DOI: 10.1097/coc.0b013e31819cccaf] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate whether the incidence of thromboembolic events (venous and arterial) increases when bevacizumab-based chemotherapy and erythropoietin stimulating agents (ESAs) are used in combination versus alone. METHODS A retrospective, pilot study of 79 colorectal cancer patients treated with chemotherapy were divided into 3 groups: bevacizumab (n = 28), ESA (n = 21), and bevacizumab plus ESA (n = 28). The primary end point was the incidence of thromboembolic events. Secondary endpoints included median time-to-event; effect of anticoagulation; and association with concurrent chemotherapy, baseline risk factors, hemoglobin, and performance status. RESULTS The incidence of thromboembolic events was 11% in the bevacizumab group, 23.8% in the ESA group, and 30% in the combination group (P = 0.194). The median time-to-event was 7.5, 3.5, and 2.5 months, respectively (P = 0.060). The 5 month difference in time-to-event between the bevacizumab group and combination group was significant (P = 0.045). When combining all patients, ESA treatment, prior venous thromboembolic event (VTE), obesity, cardiac disease, and use of exogenous hormones were strong predictors for thromboembolic events. Prior VTE was a strong predictor in those patients in the combination group. CONCLUSION The incidence of thromboembolic events was increased with the combination of bevacizumab plus ESA compared with either agent alone with chemotherapy. Median time-to-event in the combination group was significantly shorter compared with the bevacizumab group. Prior VTE, cardiac disease, obesity, and exogenous hormone use should be taken in consideration when using the combination of bevacizumab and ESAs.
Collapse
|
206
|
Barbera L, Thomas G. Erythropoiesis stimulating agents, thrombosis and cancer. Radiother Oncol 2010; 95:269-76. [PMID: 20219259 DOI: 10.1016/j.radonc.2010.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 02/09/2010] [Indexed: 12/27/2022]
Abstract
Venous thromboembolism (VTE) is common in cancer and is associated with both morbidity and mortality. Erythropoiesis stimulating agents (ESAs) were originally developed to correct anemia. Recent trials in cancer patients however, raise concerns over both increased VTE rates and the possibility of worse tumour outcomes and increased mortality with ESA use. The most common reason offered for explaining the possible negative impact of ESAs on cancer outcomes has been the stimulation of erythropoietin receptors on tumour cells. Despite an extensive literature, it is unlikely that most practicing appreciate the intricate relationship and interaction between the coagulation pathways, angiogenesis and tumour progression and ESA effects. This paper will review these connections and interactions and examine the hypothesis that other mechanisms may underlie the possible negative impact of ESAs on cancer outcomes.
Collapse
Affiliation(s)
- Lisa Barbera
- Department of Radiation Oncology, University of Toronto, Canada
| | | |
Collapse
|
207
|
|
208
|
Mikhael J, Melosky B, Cripps C, Rayson D, Kouroukis CT. Canadian supportive care recommendations for the management of anemia in patients with cancer. ACTA ACUST UNITED AC 2010; 14:209-17. [PMID: 17938704 PMCID: PMC2002484 DOI: 10.3747/co.2007.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Anemia is a common finding in cancer patients, most often as a result of chemotherapy. Management of anemia requires a comprehensive approach of appropriate diagnosis, exclusion of reversible causes, use of erythropoiesis-stimulating agents (esas), and iron supplementation. Recently, consensus guidelines on the management of chemotherapy-induced anemia were published in Europe and the United States. The present review is intended to be a practical guide for Canadian physicians, based on published guidelines, but specifically tailored to the Canadian environment. Recommendations for the use of esas are presented, including initiation, target hemoglobin, dosing and adjustments, monitoring, and re-initiation. Issues of safety are also addressed, including thromboembolic risk, impact on survival, and tumour progression. The importance of iron metabolism and the use of iron supplementation (both oral and parenteral) is discussed.
Collapse
Affiliation(s)
- J Mikhael
- Princess Margaret Hospital, Toronto, Ontario.
| | | | | | | | | |
Collapse
|
209
|
Vogiatzi G, Briasoulis A, Tousoulis D, Papageorgiou N, Stefanadis C. Is there a role for erythropoietin in cardiovascular disease? Expert Opin Biol Ther 2010; 10:251-264. [PMID: 20028188 DOI: 10.1517/14712590903547819] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IMPORTANCE OF THE FIELD Despite the advances in the cardiovascular field, cardiovascular diseases remain an important health problem with a high mortality rate. Novel therapeutic attempts that target myocardial ischemia and heart failure offer attractive adjuncts and/or alternatives to commonly employed regimens. The development of novel laboratory technologies over the last decade has led to substantial progress in bringing new therapies to the bedside. AREAS COVERED IN THIS REVIEW Current experimental and clinical trials in the use of erythropoietin (EPO) in cardiovascular diseases are reviewed. WHAT THE READER WILL GAIN This review will widen knowledge of the therapeutic potential of EPO's non-erythropoietic beneficial effects in a clinical cardiovascular setting. TAKE HOME MESSAGE Results from preclinical trials regarding the non-erythropoietic effects of erythropoietin are really encouraging. Further clinical studies are warranted to define the beneficial role of EPO in the clinical setting of coronary artery disease, heart failure and peripheral artery disease.
Collapse
Affiliation(s)
- Georgia Vogiatzi
- Athens University Medical School, Hippokration Hospital, First Cardiology Unit, Vasilissis Sofias 114, 115 28, Athens, Greece
| | | | | | | | | |
Collapse
|
210
|
Kellokumpu-Lehtinen PL, Puistola U, Paija O, Taimela E, Hirvonen O, Raassina S, Riska H. Anaemia: a rare but neglected problem among Finnish patients receiving chemotherapy for solid tumours. Support Care Cancer 2010; 19:149-53. [PMID: 20101415 DOI: 10.1007/s00520-009-0809-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 12/23/2009] [Indexed: 11/28/2022]
Abstract
GOALS OF THE WORK Anaemia is very frequently diagnosed among cancer patients. Use of erythropoietins has proved to be effective in reducing the need of transfusions and enhancing patients' quality of life, but may also have detrimental effects in treating nonanemic asymptomatic patients. We assessed the frequency of anaemia and the frequency with which it was diagnosed and treated in different types of solid tumours treated at outpatient chemotherapy policlinics. MATERIALS AND METHODS During the study period, altogether 733 consecutive subjects received chemotherapy at the five Finnish University Hospitals. Their data were collected. The physician who was responsible for the chemotherapy treatment was unaware of the survey. The response to anaemia (treated or not, the modality of treatment) were established from patients records; 69% were females, mean age was 61 years (range, 24-92). RESULTS The median haemoglobin level was 12.7 g/dL (range, 8.9-15.5 g/dL). About one third of the patients (200/733, 27%) had a value less than 12 g/dL. In only 15% of these cases was there any documentation of response or a possible treatment option for anaemia. On the other hand, only 12% of all patients (N=91) had a haemoglobin value less than 11 g/dL. However, in most of them anaemia had not been considered; in only 25% of cases was an active treatment option selected. CONCLUSIONS According to our survey, anaemia was less common in our patients than in the European Cancer Anaemia Survey. Only a minority of chemotherapy patients receiving their treatments as outpatients would need active treatment for their anaemia.
Collapse
|
211
|
Ngo K, Kotecha D, Walters JA, Manzano L, Palazzuoli A, van Veldhuisen DJ, Flather M. Erythropoiesis-stimulating agents for anaemia in chronic heart failure patients. Cochrane Database Syst Rev 2010:CD007613. [PMID: 20091643 DOI: 10.1002/14651858.cd007613.pub2] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Chronic heart failure (CHF) is a leading cause of morbidity and mortality worldwide. Anaemia is a common (12-55%) co-morbid condition and is associated with worsening symptoms and increased mortality. Anaemia is treatable and can be targeted in the treatment of patients with CHF. Erythropoiesis-stimulating agents (ESA), supplemented by iron therapy, are used to treat anaemia in chronic kidney disease and cancer, however safety concerns have been raised in these patients. The clinical benefit and safety of these agents in CHF remains unclear. OBJECTIVES To assess the benefits and risks of ESA for CHF patients with anaemia. SEARCH STRATEGY We searched the Cochrane Central Register of Controlled Trials (The Cochrane Library 2008, Issue 3), MEDLINE (1950 to October 2008), EMBASE (1980 to October 2008) and reference lists of articles. No language restrictions were applied. SELECTION CRITERIA Randomised controlled trials of any ESA, with or without iron therapy, in CHF patients were eligible for inclusion. DATA COLLECTION AND ANALYSIS Three reviewers independently assessed study quality and extracted data. Original authors were contacted for additional information. The outcomes of interest were: exercise tolerance, haemoglobin level, New York Heart Association (NYHA) functional class, quality of life, left-ventricular ejection fraction, B-type natriuretic peptide, CHF-related hospitalisations, all-cause mortality and adverse effects. Risk ratios (RR) were calculated for dichotomous data and weighted mean difference (WMD) for continuous data. MAIN RESULTS Eleven studies (794 participants) were included. Overall quality of studies was moderate with nine studies being placebo-controlled but only five double-blinded. Compared to control, ESA treatment significantly improved exercise duration by 96.8 seconds (95% CI 5.2 to 188.4, p=0.04) and 6-minute walk distance by 69.3 metres (95% CI 17.0 to 121.7, p=0.009). Benefit was also noted in terms of peak VO2 (+2.29 mL/kg/min, p=0.007), NYHA class (-0.73, p<0.001), ejection fraction (+5.8%, p<0.001), B-type natriuretic peptide (-226.99 pg/mL, p<0.001) and quality-of-life indicators, with a mean increase in haemoglobin of 1.98 g/dL (p<0.0001). There was also a significantly lower rate of heart failure related hospitalisations (RR 0.62, 95% CI 0.44 to 0.87) and lower all-cause mortality (RR 0.61, 95% CI 0.37 to 0.99). No increase in adverse events with ESA therapy was observed, however studies were of small sample sizes and limited duration. AUTHORS' CONCLUSIONS Meta-analysis of small RCTs suggests that ESA treatment in patients with symptomatic CHF and mild anaemia (haemoglobin more than 10g/dL) can improve anaemia and exercise tolerance, reduce symptoms and have benefits on clinical outcomes. Confirmation requires well-designed studies with careful attention to dose, haemoglobin treatment target and associated iron therapy.
Collapse
Affiliation(s)
- Katherine Ngo
- School of Medicine, University of Tasmania, 43 Collins Street, Hobart, Tasmania, Australia, 7005
| | | | | | | | | | | | | |
Collapse
|
212
|
Glaspy J, Crawford J, Vansteenkiste J, Henry D, Rao S, Bowers P, Berlin JA, Tomita D, Bridges K, Ludwig H. Erythropoiesis-stimulating agents in oncology: a study-level meta-analysis of survival and other safety outcomes. Br J Cancer 2010; 102:301-15. [PMID: 20051958 PMCID: PMC2816662 DOI: 10.1038/sj.bjc.6605498] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND: Cancer patients often develop the potentially debilitating condition of anaemia. Numerous controlled studies indicate that erythropoiesis-stimulating agents (ESAs) can raise haemoglobin levels and reduce transfusion requirements in anaemic cancer patients receiving chemotherapy. To evaluate recent safety concerns regarding ESAs, we carried out a meta-analysis of controlled ESA oncology trials to examine whether ESA use affects survival, disease progression and risk of venous-thromboembolic events. METHODS: This meta-analysis included studies from the 2006 Cochrane meta-analysis, studies published/updated since the 2006 Cochrane report, and unpublished trial data from Amgen and Centocor Ortho Biotech. The 60 studies analysed (15 323 patients) were conducted in the settings of chemotherapy/radiochemotherapy, radiotherapy only treatment or anaemia of cancer. Data were summarised using odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS: Results indicated that ESA use did not significantly affect mortality (60 studies: OR=1.06; 95% CI: 0.97–1.15) or disease progression (26 studies: OR=1.01; 95% CI: 0.90–1.14), but increased the risk for venous-thromoboembolic events (44 studies: OR=1.48; 95% CI: 1.28–1.72). CONCLUSION: Though this meta-analysis showed no significant effect of ESAs on survival or disease progression, prospectively designed, future randomised clinical trials will further examine the safety and efficacy of ESAs when used according to the revised labelling information.
Collapse
Affiliation(s)
- J Glaspy
- Department of Medicine-Hematology and Oncology, David Geffen School of Medicine at University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 550, Los Angeles, CA 90095-6996 USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
|
214
|
Henry DH. Parenteral iron therapy in cancer-associated anemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2010; 2010:351-356. [PMID: 21239818 DOI: 10.1182/asheducation-2010.1.351] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Anemia is common in cancer patients. Its cause is multifactorial, so a brief workup is always necessary to rule out simple, reversible causes. Anemia of chronic disease/inflammation and chemotherapy-induced anemia are the most common causes. Symptomatic or clinically severe anemia may require treatment with blood transfusion or an erythropoiesis-stimulating agent (ESA). If ESA therapy is chosen, developing evidence now suggests that, similar to chronic renal failure patients on hemodialysis, the addition of intravenous iron can improve the response to ESA because of iron-restricted erythropoiesis, even in the iron-replete patient.
Collapse
Affiliation(s)
- David H Henry
- Joan Karnell Cancer Center at Pennsylvania Hospital, Philadelphia, PA 19106, USA.
| |
Collapse
|
215
|
Abstract
This article reviews the principles of systemic cancer treatment in older individuals. These include: assessment of physiologic age with a comprehensive geriatric assessment (CGA), adjustment of chemotherapy doses to the patient's renal function, and prevention of myelotoxicity with hemopoietic growth factors. Other complications that become more common with age include mucositis, peripheral neuropathy and cardiomyopathy. Two chronic complications of chemotherapy become more common with age, including myelodysplasia and chronic cardiomyopathy. The goal of systemic cancer treatment in the older person should include prolongation of active life-expectancy and compression of morbidity in addition to prolongation of survival and symptom management.
Collapse
|
216
|
Winczura P, Jassem J. Combined treatment with cytoprotective agents and radiotherapy. Cancer Treat Rev 2009; 36:268-75. [PMID: 20044209 DOI: 10.1016/j.ctrv.2009.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 11/29/2009] [Accepted: 12/02/2009] [Indexed: 10/20/2022]
Abstract
Radiotherapy is associated with several toxicities affecting healthy tissues. One of the strategies aimed at decreasing radiation toxicity is the use of radioprotective agents, such as amifostine and palifermin, or factors stimulating hemopoetic stem cells (colony stimulating factors, CSFs): granulocyte-CSF, granulocyte macrophage-CSF and recombinant erythropoetins. The potential beneficial effect of these substances demonstrated in preclinical in vitro and in vivo studies led to numerous clinical trials. This review addresses the current experience on the use of cytoprotective agents in combination with radiotherapy, with particular focus on the safety of these approaches. Despite a relatively large body of literature data, the role of cytoprotective agents combined with radiotherapy remains controversial. Overall, their use in this application is still limited due to modest radioprotective effect for normal tissues, potential risk of tumor protection and increased treatment toxicity. The use of erythropoetins in combination with radiotherapy should generally be discouraged, whereas the safe and effective application of other agents warrants further investigations.
Collapse
Affiliation(s)
- Piotr Winczura
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Poland
| | | |
Collapse
|
217
|
Abstract
Anemia is frequent in cancer patients and its incidence increases with chemotherapy. The probability of requiring transfusions also increases with chemotherapy. Anemia negatively impacts survival and accentuates fatigue in cancer patients. Cancer promotes inflammatory cytokine production, which suppresses erythropoiesis and erythropoietin (EPO) production. Erythropoiesis-stimulating agents (ESAs) improve erythropoiesis and reduce transfusion needs in anemic cancer patients receiving chemotherapy. However, meta-analyses have shown an increased risk of thromboembolic (TE) events with ESA use during chemotherapy, but not increased on-study mortality or reduced overall survival. Three reasons have been proposed to explain why ESAs might have adverse effects in anemic cancer patients: tumor progression due to stimulation of tumor cell EPO receptors; increased risk of TE; and reduced survival. However, erythropoietin is not an oncogene, nor is the EPO receptor. It has also been demonstrated that erythropoietin does not stimulate tumor proliferation. Increased TE risk associated with ESAs is probably a consequence of increased blood viscosity due to excessive RBC mass elevation with concomitant plasma volume contraction, nitric oxide scavenging, and endothelial cell activation. Increased ESA dosing may also impact survival negatively because EPO contracts the plasma volume and stimulates inflammatory cytokine production independently of increasing erythropoiesis. Furthermore, transfusions themselves are associated with an increase in TE and plasma volume contraction, and these events are potentiated when ESAs are given with transfusions. An update on the management of anemia in oncology, the potential adverse events of ESAs, the benefits and risks of transfusions, and QoL are discussed in this paper.
Collapse
Affiliation(s)
- Jerry L Spivak
- Johns Hopkins University School of Medicine, Baltimore, Maryland 21210, USA.
| | | | | |
Collapse
|
218
|
Aapro M, Spivak JL. Update on erythropoiesis-stimulating agents and clinical trials in oncology. Oncologist 2009; 14 Suppl 1:6-15. [PMID: 19762512 DOI: 10.1634/theoncologist.2009-s1-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Anemia commonly occurs among cancer patients receiving chemotherapy. In these patients, erythropoiesis-stimulating agents (ESAs) are effective in managing anemia but there is an increased risk for thrombovascular events. In more recent randomized clinical trials, there have been differing results regarding the impact of ESAs on overall survival and mortality. The balance between studies that show higher ESA-associated mortality and those that don't show ESA-associated mortality is examined in this review. This review discusses where we stand today on anemia management in cancer patients. Preliminary results from a recent independent patient data meta-analysis for on-study deaths and overall survival in patients receiving chemotherapy (the only oncology population for which ESA treatment is currently indicated) showed no statistically significant difference between the ESA and control groups (on-study deaths hazard ratio [HR], 1.10; 95% confidence interval [CI], 0.98-1.24; overall survival HR, 1.04; 95% CI, 0.97-1.11, compared with controls). Possible factors that could influence study results are discussed in this review. There are no convincing data to support ESA-induced tumor stimulation in patients. ESAs decrease RBC transfusion needs and sustain targeted hemoglobin levels, and this ESA response does not significantly impact overall survival or mortality when ESAs are used within guidelines and labeling. However, based on the currently available data and meta-analysis, the use of ESAs has to be carefully balanced against any possible risk for higher mortality.
Collapse
Affiliation(s)
- Matti Aapro
- Multidisciplinary Oncology Institute, Genolier, Switzerland.
| | | |
Collapse
|
219
|
Abstract
Venous thromboembolism (VTE) is a frequent complication of cancer and cancer treatment and is associated with multiple clinical consequences, including recurrent VTE, bleeding, and an increase in the risk of death. Although the risks associated with VTE have been well recognized in surgical cancer patients, there is also considerable and increasing risk in medical cancer patients. VTE risk factors in medical cancer patients include the type and stage of cancer, major comorbid illnesses, current hospitalization, active chemotherapy, hormone therapy, and antiangiogenic agents. Low-molecular-weight heparins (LMWHs) are recommended commonly for the prevention of VTE in hospitalized cancer patients and in higher risk ambulatory cancer patients because of their favorable risk-to-benefit profile. These agents have demonstrated effectiveness in both the primary and secondary prevention of VTE in medical cancer patients. Extended-duration anticoagulant therapy is often recommended to reduce the risk of VTE recurrence in patients with cancer. LMWHs are often used for long-term prophylaxis because of a reduced need for coagulation monitoring, few major bleeding episodes, and once-daily dosing. Despite clinical and practical benefits, a substantial proportion of medical cancer patients do not receive VTE prophylaxis. To improve the appropriate prevention and treatment of VTE in cancer patients, guidelines have been published recently by the American Society of Clinical Oncology and the National Comprehensive Cancer Network. Widespread dissemination and application of these guidelines are encouraged to improve the appropriate use of these agents and to improve clinical outcomes in medical cancer patients at risk for VTE and its complications.
Collapse
Affiliation(s)
- Gary H Lyman
- Department of Medicine, Duke University School of Medicine and the Duke Comprehensive Cancer Center, Durham, North Carolina 27710, USA.
| |
Collapse
|
220
|
Spanish Society of Medical Oncology consensus on the use of erythropoietic stimulating agents in anaemic cancer patients. Clin Transl Oncol 2009; 11:727-36. [DOI: 10.1007/s12094-009-0435-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
221
|
Effects of recombinant human erythropoietin on platelet activation in acute myocardial infarction: results of a double-blind, placebo-controlled, randomized trial. Am Heart J 2009; 158:941-7. [PMID: 19958860 DOI: 10.1016/j.ahj.2009.06.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 06/20/2009] [Indexed: 02/06/2023]
Abstract
BACKGROUND Erythropoietin mitigates myocardial damage and improves ventricular performance after experimental ischemic injury. This study assessed safety and efficacy markers relevant to the biological activity of recombinant human erythropoietin (rHuEpo) in patients with acute myocardial infarction (MI). METHODS We conducted a prospective, placebo-controlled, randomized, double-blind trial to determine the effects of intravenous rHuEpo (200 U/kg daily for 3 consecutive days) on measures of platelet and endothelial cell activation, soluble Fas ligand, and peripheral blood mononuclear cell (PBMC) expression of angiogenesis signaling proteins in 44 subjects with acute MI treated with aspirin and clopidogrel after successful percutaneous coronary intervention. RESULTS Recombinant human erythropoietin did not alter bleeding time, platelet function assay closure time, von Willebrand factor levels, soluble P-selectin, or soluble Fas ligand levels when compared with placebo. By contrast, rHuEpo significantly increased expression of erythropoietin receptor, vascular endothelial growth factor receptor Flt-1, and phosphorylated phosphatidylinositol 3-kinase in PBMCs when compared with placebo (all Ps < .05). CONCLUSIONS In acute MI patients treated with aspirin and clopidogrel, short-term administration of rHuEpo did not alter markers of platelet and endothelial cell activation associated with thrombosis, yet did increase expression of angiogenesis signaling proteins in PBMCs when compared with placebo. These data provide preliminary evidence of safety and biologic activity of rHuEpo at this dosing and support continued enrollment in ongoing efficacy trials.
Collapse
|
222
|
Lüthi F, Pless M, Leyvraz S, Biedermann B, Müller E, Hermann R, Monnerat C. Dose reduction of epoetin-alpha in the prevention of chemotherapy-induced anaemia. Support Care Cancer 2009; 18:1515-20. [PMID: 19921283 DOI: 10.1007/s00520-009-0773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Accepted: 10/26/2009] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Anaemia during chemotherapy is often left untreated. Erythropoiesis-stimulating agents are frequently used to treat overt anaemia. Their prophylactic use, however, remains controversial and raises concerns about cost-effectiveness. Therefore, we assessed the efficacy of a dose-reduction schedule in anaemia prophylaxis. MATERIALS AND METHODS The study included patients with untreated solid tumours about to receive platinum-based chemotherapy and had haemoglobin (Hb) levels ≥11 g/dL. Epoetin-α was administered at a dose level of 3 × 10,000 U weekly as soon as Hb descended to < 13 g/dL. Dose reductions to 3 × 4,000 U and 3 × 2,000 U weekly were planned in 4-week intervals if Hb stabilised in the range of 11-13 g/dL. Upon ascending to ≥13 g/dL, epoetin was discontinued. Iron supplements of 100 mg intravenous doses were given weekly. Of 37 patients who enrolled, 33 could be evaluated. RESULTS AND DISCUSSION Their median Hb level was 13.7 (10.9-16.2) g/dL at baseline and descended to 11.0 (7.4-13.8) g/dL by the end of chemotherapy. Anaemia (Hb < 10 g/dL) was prevented in 24 patients (73%). The mean dose requirement for epoetin-α was 3 × 5,866 U per week per patient, representing a dose reduction of 41%. Treatment failed in nine patients (27%), in part due to epoetin-α resistance in four (12%) and blood transfusion in three (9%) patients. CONCLUSION Dose reduction was as effective as fixed doses in anaemia prophylaxis but reduced the amount of prescribed epoetin substantially.
Collapse
Affiliation(s)
- François Lüthi
- Centre Pluridisciplinaire d'Oncologie, University Hospital-CHUV BH06, Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
223
|
Bell D, Grimes D, Gurney H, Dalley D, Blackwell T, Fox R, Jeffery M. Outcomes and predicting response in anaemic chemotherapy patients treated with epoetin alfa. A multicentre, 4-month, open-label study in Australia and New Zealand. Intern Med J 2009; 38:751-7. [PMID: 19143877 DOI: 10.1111/j.1445-5994.2008.01736.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The aim of the study was to evaluate the effectiveness, safety, and clinical outcomes of erythropoietin therapy in the treatment of anaemic cancer subjects receiving chemotherapy and to examine hypochromic red blood cell measurement as an indicator of functional iron sufficiency and as a predictor of responsiveness or non-responsiveness to erythropoietin therapy. METHODS Patients who had a non-myeloid malignancy, had Hb < or = 11.0 g/dL, had a life expectancy of more than 6 months, were 18 years or older, were receiving chemotherapy and would continue to be treated for at least 2 months were given s.c. epoetin alfa three times a week. RESULTS Haemoglobin levels increased significantly at all time periods compared with baseline and the number of transfusions received decreased significantly at all time periods compared with baseline. Quality of life as measured by Functional Assessment of Cancer Therapy-Anaemia showed significant increases at months 2 and 4 and there were significant improvements in the fatigue subscale at both time points (P < 0.05). Significant improvements at end-point were observed for the physical, emotional and functional well-being, and additional concern subscales (all P < 0.05). Haematocrit and reticulocytes increased significantly at end-point compared with at baseline (haematocrit 33.4 vs 28.3%, P < 0.001; reticulocytes 105.8 vs 78.6 x 10(9)/dL, P = 0.005). The percentage of hypochromic red blood cells did not show predictive value for response to treatment status. CONCLUSION Epoetin alfa improved haemoglobin levels and quality of life in anaemic cancer patients receiving chemotherapy.
Collapse
Affiliation(s)
- D Bell
- Department of Clinical Oncology, Royal North Shore Hospital, Sydney, St Leonards, NSW 2065, Australia.
| | | | | | | | | | | | | |
Collapse
|
224
|
Hershman DL, Buono DL, Malin J, McBride R, Tsai WY, Neugut AI. Patterns of use and risks associated with erythropoiesis-stimulating agents among Medicare patients with cancer. J Natl Cancer Inst 2009; 101:1633-41. [PMID: 19903808 DOI: 10.1093/jnci/djp387] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Erythropoiesis-stimulating agents (erythropoietin and darbepoietin) have been approved to reduce the number of blood transfusions required during chemotherapy; however, concerns about the risks of venous thromboembolism and mortality exist. METHODS We identified patients who were aged 65 years or older in the Surveillance, Epidemiology, and End Results-Medicare database; who were diagnosed with colon, non-small cell lung, or breast cancer or with diffuse large B-cell lymphoma from January 1, 1991, through December 31, 2002; and who received chemotherapy. The main outcome measures were claims for use of an erythropoiesis-stimulating agent, blood transfusion, venous thromboembolism (ie, deep vein thrombosis or pulmonary embolism), and overall survival. We used multivariable logistic regression models to analyze the association of erythropoiesis-stimulating agent use with clinical and demographic variables. We used time-dependent Cox proportional hazards models to analyze the association of time to receipt of first erythropoiesis-stimulating agent with venous thromboembolism and overall survival. All statistical tests were two-sided. RESULTS Among 56,210 patients treated with chemotherapy from 1991 through 2002, 15,346 (27%) received an erythropoiesis-stimulating agent. The proportion of patients receiving erythropoiesis-stimulating agents increased from 4.8% in 1991 to 45.9% in 2002 (P < .001). Use was associated with more recent diagnosis, younger age, urban residence, comorbidities, receipt of radiation therapy, female sex, and metastatic or recurrent cancer. The rate of blood transfusion per year during 1991-2002 remained constant at 22%. Venous thromboembolism developed in 1796 (14.3%) of the 12,522 patients who received erythropoiesis-stimulating agent and 3400 (9.8%) of the 34,820 patients who did not (hazard ratio = 1.93, 95% confidence interval = 1.79 to 2.07). Overall survival was similar in both groups. CONCLUSION Use of erythropoiesis-stimulating agent increased rapidly after its approval in 1991, but the blood transfusion rate did not change. Use of erythropoiesis-stimulating agents was associated with an increased risk of venous thromboembolism but not of mortality.
Collapse
Affiliation(s)
- Dawn L Hershman
- Columbia University Medical Center, 161 Fort Washington Ave, 10-1068, New York, NY 10032, USA.
| | | | | | | | | | | |
Collapse
|
225
|
|
226
|
Mees G, Fonteyne P, Ceelen W, Boterberg T, Pauwels P, Vangestel C, Van Damme N, Peeters M, Dierckx R, Van De Wiele C. Combined effect of EPO and radiotherapy on the expression of endogenous molecular markers of tumor metabolism and metastasis. Cancer Biother Radiopharm 2009; 24:565-72. [PMID: 19877886 DOI: 10.1089/cbr.2009.0621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Erythropoietin (EPO) has been used to correct cancer-related anemia and to improve tumor hypoxia, which both adversely affect the clinical condition of cancer patients and response to radiotherapy. Data available on the effects of EPO treatment in cancer are, however, conflicting. Several clinical studies investigating the influence of EPO treatment have given contradictory results as to whether or not this treatment positively influences survival. In light of these conflicting results, we studied the effects of EPO treatment either alone or in combination with radiotherapy on tumor oxygenation and on the expression pattern of several proteins related to tumor metabolism, survival, and spread in a rat colorectal cancer model. We found a statistically significant upregulation of hexokinase I, N-cadherin, and glucose transporter 3 when EPO treatment was combined with radiotherapy. Because these three proteins have distinct functions in protecting the cell in compromised conditions, these results indicate a detrimental role for the combination of EPO treatment and radiotherapy through the stimulation of tumor-cell metabolism, inhibition of apoptosis, and stimulation of tumor spread and seem to indicate that recombinant human EPO treatment negatively modulates radiotherapy efficacy.
Collapse
Affiliation(s)
- Gilles Mees
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Klapholz M, Abraham WT, Ghali JK, Ponikowski P, Anker SD, Knusel B, Sun Y, Wasserman SM, van Veldhuisen DJ. The safety and tolerability of darbepoetin alfa in patients with anaemia and symptomatic heart failure. Eur J Heart Fail 2009; 11:1071-7. [DOI: 10.1093/eurjhf/hfp130] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Marc Klapholz
- Department of Cardiology; New Jersey Medical School; Newark NJ USA
| | - William T. Abraham
- Division of Cardiovascular Medicine; The Ohio State University; Columbus OH USA
| | - Jalal K. Ghali
- Detroit Medical Center; Cardiovascular Institute; Detroit MI USA
| | - Piotr Ponikowski
- Faculty of Public Health, Cardiac Department; Medical University; Wroclaw Poland
- Cardiac Department; Military Hospital; Wroclaw Poland
| | - Stefan D. Anker
- Division of Applied Cachexia Research, Department of Cardiology; Charité Campus Virchow-Klinikum; Berlin Germany
- Centre for Clinical and Basic Research; IRCCS San Raffaele; Rome Italy
| | | | - Yan Sun
- Amgen, Inc.; Thousand Oaks CA USA
| | | | - Dirk J. van Veldhuisen
- Department of Cardiology; University Medical Centre Groningen; Groningen The Netherlands
| |
Collapse
|
228
|
Khorana AA, Connolly GC. Assessing risk of venous thromboembolism in the patient with cancer. J Clin Oncol 2009; 27:4839-47. [PMID: 19720906 PMCID: PMC2764392 DOI: 10.1200/jco.2009.22.3271] [Citation(s) in RCA: 382] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2009] [Accepted: 06/23/2009] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Patients with cancer are increasingly at risk for venous thromboembolism (VTE). Rates of VTE, however, vary markedly among patients with cancer. DESIGN This review focuses on recent data derived from population-based, hospital-based, and outpatient cohort studies of patients with cancer that have identified multiple clinical risk factors as well as candidate laboratory biomarkers predictive of VTE. RESULTS Clinical risk factors for cancer-associated VTE include primary tumor site, stage, initial period after diagnosis, presence and number of comorbidities, and treatment modalities including systemic chemotherapy, antiangiogenic therapy, and hospitalization. Candidate predictive biomarkers include elevated platelet or leukocyte counts, tissue factor, soluble P-selectin, and D-dimer. A recently validated risk model, incorporating some of these factors, can help differentiate patients at high or low risk for developing VTE while receiving chemotherapy. CONCLUSION Identifying patients with cancer who are most at risk for VTE is essential to better target thromboprophylaxis, with the eventual goal of reducing the burden as well as the consequences of VTE for patients with cancer.
Collapse
Affiliation(s)
- Alok A Khorana
- James P. Wilmot Cancer Center, and Department of Medicine, University of Rochester, Rochester, NY, USA.
| | | |
Collapse
|
229
|
Affiliation(s)
- Gary H Lyman
- Duke University and Duke Comprehensive Cancer Center, Durham, NC, USA
| | | |
Collapse
|
230
|
Doyle MK, Rahman MU, Han C, Han J, Giles J, Bingham CO, Bathon J. Treatment with infliximab plus methotrexate improves anemia in patients with rheumatoid arthritis independent of improvement in other clinical outcome measures-a pooled analysis from three large, multicenter, double-blind, randomized clinical trials. Semin Arthritis Rheum 2009; 39:123-31. [PMID: 18823645 DOI: 10.1016/j.semarthrit.2008.08.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 06/27/2008] [Accepted: 08/02/2008] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To evaluate the effect of antitumor necrosis factor-alpha monoclonal antibody infliximab treatment on anemia in patients with rheumatoid arthritis (RA). METHODS Data from patients with RA who received infliximab or placebo in the multicenter, placebo-controlled, double-blind, randomized ATTRACT, ASPIRE, and START studies were included in this post-hoc, pooled analysis. Infliximab (3 to 10 mg/kg) was administered every 4 or 8 weeks, and all patients received stable doses of methotrexate (MTX). We determined the percentage of anemic patients (baseline hemoglobin level <12 g/dL) who had an increase from baseline in hemoglobin level greater than or equal to 1 or 2 g/dL or achieved normal hemoglobin level at week 22. The association of improvement in anemia with improvement in clinical parameters was also evaluated. RESULTS Among patients with anemia at baseline, infliximab plus MTX treatment produced a significantly greater mean (standard deviation) increase in hemoglobin level from baseline to week 22 (0.74 [1.12], P < 0.0001) than placebo plus MTX (0.30 [0.92]). Significantly (P < 0.001) greater proportions of anemic patients treated with infliximab plus MTX had either at least a 1 g/dL (40%) or at least a 2 g/dL (12%) increase in hemoglobin level from baseline to week 22 or achieved normal hemoglobin level (43%) when compared with placebo plus MTX (19, 5, and 28%, respectively). Greater improvement in hemoglobin level among infliximab plus MTX-treated patients was consistently observed across subgroups and in patients without clinical response (American College of Rheumatology 20% response criteria) at week 22. Multiple regression analysis indicated that the effect of infliximab plus MTX on anemia was independent of improvement in disease activity. CONCLUSION Treatment with infliximab plus MTX significantly improved hemoglobin level among anemic RA patients when compared with treatment with placebo plus MTX, even after adjusting for improvement in disease activity.
Collapse
Affiliation(s)
- Mittie K Doyle
- Immunology, Clinical Research and Development, Centocor Inc., Malvern, PA 19355, USA.
| | | | | | | | | | | | | |
Collapse
|
231
|
Abstract
Therapy with erythropoiesis-stimulating agents (ESAs) is associated with well-documented benefits to anemic cancer patients undergoing chemotherapy, most importantly a reduction in the likelihood of needing red cell transfusions. One challenge in supportive cancer care is a relative resistance to ESAs, requiring high doses with a significant rate of nonresponse. Recent advances in our understanding of iron metabolism in patients with chronic illness and the results of clinical trials indicate that parenteral iron improves ESA response in this setting. Another issue is the safety of ESA treatment in cancer patients. There is an increased risk of venous thrombosis that must be considered in clinical decision making. There are also recent data raising concerns that ESAs may enhance tumor progression or decrease patient survival. Although the preponderance of the data suggests that ESAs do not alter survival when used to treat chemotherapy-induced anemia, large well-controlled trials addressing this issue are needed.
Collapse
Affiliation(s)
- John A Glaspy
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of California, Los Angeles, California 90095, USA.
| |
Collapse
|
232
|
Vansteenkiste J, Hedenus M, Gascon P, Bokemeyer C, Ludwig H, Vermorken J, Hamilton L, Bridges K, Pujol B. Darbepoetin alfa for treating chemotherapy-induced anemia in patients with a baseline hemoglobin level < 10 g/dL versus > or = 10 g/dL: an exploratory analysis from a randomized, double-blind, active-controlled trial. BMC Cancer 2009; 9:311. [PMID: 19728887 PMCID: PMC2744706 DOI: 10.1186/1471-2407-9-311] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 09/03/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Several studies have shown that darbepoetin alfa, an erythropoiesis-stimulating agent (ESA), can reduce transfusions and increase hemoglobin (Hb) levels in patients with chemotherapy-induced anemia (CIA). Recent safety concerns, however, have prompted changes to ESA product information. In the European Union and United States, ESA therapy initiation for CIA is now recommended at a Hb level < or = 10 g/dL. The present exploratory analysis examined how ESA initiation at this Hb level may impact patient care. METHODS Data from a phase 3 randomized trial were retrospectively reanalyzed. CIA patients with nonmyeloid malignancies were randomized 1:1 to 500 mcg darbepoetin alfa every three weeks (Q3W) or 2.25 mcg/kg darbepoetin alfa weekly (QW) for 15 weeks. A previously published report from this trial showed Q3W dosing was non-inferior to QW dosing for reducing transfusions from week 5 to end-of-the-treatment period (EOTP). In the present analysis, outcomes were reanalyzed by baseline Hb <10 g/dL and > or = 10 g/dL. Endpoints included transfusion rates, Hb outcomes, and safety profiles. RESULTS This study reanalyzed 351 and 354 patients who initiated ESA therapy at a baseline Hb of <10 g/dL or > or = 10 g/dL, respectively. From week 5 to EOTP, the estimated Kaplan-Meier transfusion incidence (Q3W vs QW) was lower in the > or = 10 g/dL baseline-Hb group (14% vs 21%) compared with the <10 g/dL baseline-Hb group (36% vs 41%). By week 5, the > or = 10 g/dL baseline-Hb group, but not the <10 g/dL baseline-Hb group, achieved a mean Hb > or = 11 g/dL. The Kaplan-Meier estimate of percentage of patients (Q3W vs QW) who achieved Hb > or = 11 g/dL from week 1 to EOTP was 90% vs 85% in the > or = 10 g/dL baseline-Hb group and 54% vs 57% in the <10 g/dL baseline-Hb group. Both baseline-Hb groups maintained mean Hb levels <12 g/dL and had similar safety profiles, though more patients in the > or = 10 g/dL baseline-Hb group reached the threshold Hb of > or = 13 g/dL. CONCLUSION In this exploratory analysis, darbepoetin alfa Q3W and QW raised Hb levels and maintained mean Hb at <12 g/dL in both baseline-Hb groups. The > or = 10 g/dL baseline-Hb group had fewer transfusions and faster anemia correction. Additional studies should prospectively evaluate the relationship between Hb levels at ESA initiation and outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier NCT00118638.
Collapse
Affiliation(s)
- Johan Vansteenkiste
- Respiratory Oncology Unit (Pulmonology), University Hospital Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Katodritou E, Dimopoulos MA, Zervas K, Terpos E. Update on the use of erythropoiesis-stimulating agents (ESAs) for the management of anemia of multiple myeloma and lymphoma. Cancer Treat Rev 2009; 35:738-43. [PMID: 19733008 DOI: 10.1016/j.ctrv.2009.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Revised: 08/04/2009] [Accepted: 08/06/2009] [Indexed: 11/20/2022]
Abstract
Anemia is a common side-effect of patients with multiple myeloma (MM) and lymphoma. The etiology is complex, but the main cause is the underlying mechanism of anemia of chronic disease, which is characterized among others, by impairment of iron metabolism and consequently iron restricted erythropoiesis (IRE), resulting from the up-regulation of the iron distributing regulator, hepcidin. Erythopoiesis-stimulating agents (ESAs) have been the standard of care since early 90's offering high response rates and improving the quality of life of the patients. However, the role of ESAs in the treatment of cancer-related anemia has been questioned recently, due to the growing evidence which support that ESAs may be associated with increased risk for thrombosis and may have a detrimental impact on patients' survival. Under the light of the recent considerations, the place of ESAs in the management of cancer-related anemia has been reassigned. Regarding the management of anemia in MM or lymphoma, the updated American Society of Clinical Oncology/American Society of Hematology (ASCO/ASH) 2007 clinical practice guidelines on the use of ESAs in cancer-related anemia, recommended that ESAs should be preferably omitted in patients planned to receive chemotherapy and applied in case that anemia does not improve over treatment. The quest for reliable predictors for response to ESAs and for indicators of IRE which plays a major etiological role for the development of anemia of cancer still remains an open issue. In the current review we present an update on ESAs use in anemia of MM and lymphoma.
Collapse
Affiliation(s)
- Eirini Katodritou
- Department of Hematology, Theagenion Cancer Center, Thessaloniki, Greece.
| | | | | | | |
Collapse
|
234
|
Hadland BK, Longmore GD. Erythroid-Stimulating Agents in Cancer Therapy: Potential Dangers and Biologic Mechanisms. J Clin Oncol 2009; 27:4217-26. [DOI: 10.1200/jco.2008.21.6945] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Erythropoietin-stimulating agents (ESAs) were originally designed to replace endogenous erythropoietin in patients with anemia secondary to renal failure. Their use has subsequently been expanded to include patients with anemia of other causes, including cancer patients, in whom deficiency of erythropoietin, per se, is not the primary cause of anemia. Although early studies showed promise of ESA administration in reducing the need for transfusions and improving the quality of life in cancer patients, several large randomized clinical trials have recently shown a potential detrimental effect of ESA administration on tumor progression and survival in these patients. These studies have called into question the safety of ESAs as supportive therapy in patients being treated for oncologic conditions. However, numerous questions remain to be addressed regarding the design of these studies, the effect of various targeted hemoglobin levels, and the potential biologic mechanisms proposed to explain promotion of tumor progression and reduced survival.
Collapse
Affiliation(s)
- Brandon K. Hadland
- From the Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; and Department of Medicine, Washington University School of Medicine, St Louis, MO
| | - Gregory D. Longmore
- From the Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; and Department of Medicine, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
235
|
Examining the Involvement of Erythropoiesis‐Stimulating Agents in Tumor Proliferation (Erythropoietin Receptors, Receptor Binding, Signal Transduction), Angiogenesis, and Venous Thromboembolic Events. Oncologist 2009; 14 Suppl 1:34-42. [DOI: 10.1634/theoncologist.2009-s1-34] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
236
|
Is Nephrology More at Ease Than Oncology with Erythropoiesis‐Stimulating Agents? Treatment Guidelines and an Update on Benefits and Risks. Oncologist 2009; 14 Suppl 1:57-62. [DOI: 10.1634/theoncologist.2009-s1-57] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
237
|
|
238
|
|
239
|
Hernandez E, Ganly P, Charu V, Dibenedetto J, Tomita D, Lillie T, Taylor K. Randomized, double-blind, placebo-controlled trial of every-3-week darbepoetin alfa 300 micrograms for treatment of chemotherapy-induced anemia. Curr Med Res Opin 2009; 25:2109-20. [PMID: 19601709 DOI: 10.1185/03007990903084164] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Darbepoetin alfa is effective in treating chemotherapy-induced anemia (CIA). Administration of subcutaneous darbepoetin alfa every 3 weeks (Q3W) could simplify treatment through synchronization with common Q3W chemotherapy regimens. We report results from a multicenter, randomized, double-blind, placebo-controlled, phase 3 trial evaluating the efficacy and safety of fixed-dose Q3W darbepoetin alfa in patients with a wide variety of tumor types who experienced CIA. RESEARCH DESIGN AND METHODS Patients aged > or = 18 years with anemia (hemoglobin <11 g/dL) being treated for nonmyeloid malignancy were randomized 1:1 to receive darbepoetin alfa 300 microg (n = 193) or placebo (n = 193) subcutaneously Q3W from weeks 1 to 13 in this 16-week study. Doses could be adjusted per prespecified rules. MAIN OUTCOME MEASURES The primary endpoint was the proportion of patients who received > or =1 red blood cell (RBC) transfusion between week 5 and the end of the treatment period (EOTP). The study also analyzed the proportions of patients achieving a hemoglobin concentration > or =11 g/dL and subsequently maintaining hemoglobin levels above 11 g/dL, and the change in hemoglobin concentration over time. RESULTS The proportion of patients requiring RBC transfusions between week 5 and EOTP was significantly lower in the darbepoetin alfa-treated group than in the placebo-treated group (24 vs. 41% of patients, a 16.3% difference, p < 0.001). There were no differences between the two treatment arms in quality-of-life measures. Cardiovascular/thromboembolic adverse events were uncommon and were not associated with increases in hemoglobin levels. Study limitations suggest caution in the interpretation of these results: transfusions, the primary endpoint, were recommended but not required if hemoglobin concentrations were < or =8.0 g/dL, and protocol deviations (primarily dosing errors) occurred in approximately one-half of the patients in both treatment groups. CONCLUSIONS In this study, fixed-dose Q3W darbepoetin alfa appeared to be well-tolerated and effective for the treatment of CIA. TRIAL REGISTRATION Study 20030232; ClinicalTrials.Gov Identifier: NCT00110955.
Collapse
|
240
|
Terrovitis JV, Anastasiou-Nana M, Kaldara E, Drakos SG, Nanas SN, Nanas JN. Anemia in heart failure: pathophysiologic insights and treatment options. Future Cardiol 2009; 5:71-81. [PMID: 19371205 DOI: 10.2217/14796678.5.1.71] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Anemia has been recognized as a very common and serious comorbidity in heart failure, with a prevalence ranging from 10 to 79%, depending on diagnostic definition, disease severity and patient characteristics. A clear association of anemia with worse prognosis has been confirmed in multiple heart failure trials. This finding has recently triggered intense scrutiny in order to identify the underlying pathophysiology and the best treatment options. Etiology is multifactorial, with iron deficiency and cytokine activation (anemia of chronic disease) playing the most important roles. Treatment is aimed at not only restoring hemoglobin values back to normal, but also at improving the patient's symptoms, functional capacity and hopefully the outcome. Iron supplementation and erythropoietin-stimulating agents have been used for this purpose, either alone or in combination. In this review, the recent advances in elucidating the mechanisms leading to anemia in the setting of heart failure are presented and the evidence supporting the use of different treatment approaches are discussed.
Collapse
Affiliation(s)
- John V Terrovitis
- 3rd Cardiology Department, University of Athens, School of Medicine, Athens, Greece.
| | | | | | | | | | | |
Collapse
|
241
|
Pirker R. Erythropoiesis-stimulating agents in patients with cancer: update on safety issues. Expert Opin Drug Saf 2009; 8:515-22. [DOI: 10.1517/14740330903158929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
242
|
Bohlius J, Schmidlin K, Brillant C, Schwarzer G, Trelle S, Seidenfeld J, Zwahlen M, Clarke MJ, Weingart O, Kluge S, Piper M, Napoli M, Rades D, Steensma D, Djulbegovic B, Fey MF, Ray‐Coquard I, Moebus V, Thomas G, Untch M, Schumacher M, Egger M, Engert A, Cochrane Haematological Malignancies Group. Erythropoietin or Darbepoetin for patients with cancer--meta-analysis based on individual patient data. Cochrane Database Syst Rev 2009; 2009:CD007303. [PMID: 19588423 PMCID: PMC7208183 DOI: 10.1002/14651858.cd007303.pub2] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Erythropoiesis-stimulating agents (ESAs) reduce anemia in cancer patients and may improve quality of life, but there are concerns that ESAs might increase mortality. OBJECTIVES Our objectives were to examine the effect of ESAs and identify factors that modify the effects of ESAs on overall survival, progression free survival, thromboembolic and cardiovascular events as well as need for transfusions and other important safety and efficacy outcomes in cancer patients. SEARCH STRATEGY We searched the Cochrane Library, Medline, Embase and conference proceedings for eligible trials. Manufacturers of ESAs were contacted to identify additional trials. SELECTION CRITERIA We included randomized controlled trials comparing epoetin or darbepoetin plus red blood cell transfusions (as necessary) versus red blood cell transfusions (as necessary) alone, to prevent or treat anemia in adult or pediatric cancer patients with or without concurrent antineoplastic therapy. DATA COLLECTION AND ANALYSIS We performed a meta-analysis of randomized controlled trials comparing epoetin alpha, epoetin beta or darbepoetin alpha plus red blood cell transfusions versus transfusion alone, for prophylaxis or therapy of anemia while or after receiving anti-cancer treatment. Patient-level data were obtained and analyzed by independent statisticians at two academic departments, using fixed-effects and random-effects meta-analysis. Analyses were according to the intention-to-treat principle. Primary endpoints were on study mortality and overall survival during the longest available follow-up, regardless of anticancer treatment, and in patients receiving chemotherapy. Tests for interactions were used to identify differences in effects of ESAs on mortality across pre-specified subgroups. The present review reports only the results for the primary endpoint. MAIN RESULTS A total of 13933 cancer patients from 53 trials were analyzed, 1530 patients died on-study and 4993 overall. ESAs increased on study mortality (combined hazard ratio [cHR] 1.17; 95% CI 1.06-1.30) and worsened overall survival (cHR 1.06; 95% CI 1.00-1.12), with little heterogeneity between trials (I(2) 0%, p=0.87 and I(2) 7.1%, p=0.33, respectively). Thirty-eight trials enrolled 10441 patients receiving chemotherapy. The cHR for on study mortality was 1.10 (95% CI 0.98-1.24) and 1.04; 95% CI 0.97-1.11) for overall survival. There was little evidence for a difference between trials of patients receiving different cancer treatments (P for interaction=0.42). AUTHORS' CONCLUSIONS ESA treatment in cancer patients increased on study mortality and worsened overall survival. For patients undergoing chemotherapy the increase was less pronounced, but an adverse effect could not be excluded.
Collapse
Affiliation(s)
- Julia Bohlius
- University of BernInstitute of Social and Preventive MedicineBernSwitzerland3012
| | - Kurt Schmidlin
- University of BernInstitute of Social and Preventive MedicineBernSwitzerland3012
| | - Corinne Brillant
- University Hospital of CologneCochrane Haematological Malignancies Group, Department I of Internal MedicineKerpener Str. 62CologneGermany50924
| | - Guido Schwarzer
- Insitute of Medical Biometry and Medical InformaticsGerman Cochrane CentreUniversity Medical Center FreiburgStefan‐Meier‐Str. 26FreiburgGermanyD‐79104
| | - Sven Trelle
- University of BernInstitute of Social and Preventive MedicineBernSwitzerland3012
| | - Jerome Seidenfeld
- American Society of Clinical OncologyDepartment of Cancer Policy and Clinical Affairs1900 Duke Street, Suite 200AlexandriaVAUSA22314
| | - Marcel Zwahlen
- University of BernInstitute of Social and Preventive MedicineBernSwitzerland3012
| | - Mike J Clarke
- UK Cochrane CentreNational Institute for Health ResearchSummertown Pavilion, Middle WayOxfordUKOX2 7LG
| | - Olaf Weingart
- University Hospital of CologneCochrane Haematological Malignancies Group, Department I of Internal MedicineKerpener Str. 62CologneGermany50924
| | - Sabine Kluge
- University Hospital of CologneCochrane Haematological Malignancies Group, Department I of Internal MedicineKerpener Str. 62CologneGermany50924
| | - Margaret Piper
- Blue Cross and Blue Shield AssociationTechnology Evaluation Center225 N Michigan AvenueChicagoILUSA60501
| | - Maryann Napoli
- Center for Medical Consumers130 Macdougal StreetNew YorkUSA10012
| | - Dirk Rades
- University HospitalDepartment of Radiation OncologyLübeckGermany
| | | | - Benjamin Djulbegovic
- Center for Evidence Based Medicine and Health Outcomes Research, University of South FloridaProfessor of Medicine and Oncology, H. Lee Moffitt Cancer CenterUSF Health Clinical Research,12901 Bruce B. Downs Boulevard, MDC02TampaFloridaUSA33612
| | - Martin F Fey
- University and Inselspital BernDepartment of Medical OncologyEffingerstraße 102BernSwitzerland3010
| | | | - Volker Moebus
- Academic Hospital Frankfurt am Main HöchstDepartment of GynecologyGotenstraße 6‐8Frankfurt a.M.Germany65929
| | - Gillian Thomas
- University of TorontoOdette Sunnybrook Cancer CentreRoxborough Street WTorontoOntarioCanadaM5R 1V1
| | - Michael Untch
- Helios Hospital Berlin‐BuchClinic for GynaecologySchwanebecker Chaussee 50BerlinGermany13125
| | - Martin Schumacher
- Institute of Medical Biometry and Medical Informatics, University Medical Center FreiburgGerman Cochrane CenterFreiburgGermany
| | - Matthias Egger
- Institute of Social and Preventive MedicineInstitute of Social MedicineFinkenhubelweg 11BernSwitzerland3012
| | - Andreas Engert
- University Hospital of CologneCochrane Haematological Malignancies Group, Department I of Internal MedicineKerpener Str. 62CologneGermany50924
| | | |
Collapse
|
243
|
Abstract
Objective. Drugs can induce almost the entire spectrum of hematologic disorders, affecting white cells, red cells, platelets, and the coagulation system. This paper aims to emphasize the broad range of drug-induced hematological syndromes and to highlight some of the newer drugs and syndromes.
Methods. Medline literature on drug-induced hematologic syndromes was reviewed. Most reports and reviews focus on individual drugs or cytopenias. Results. Drug-induced syndromes include hemolytic anemias, methemoglobinemia, red cell aplasia, sideroblastic anemia, megaloblastic anemia, polycythemia, aplastic anemia, leukocytosis, neutropenia, eosinophilia, immune thrombocytopenia, microangiopathic syndromes, hypercoagulability, hypoprothrombinemia, circulating anticoagulants, myelodysplasia, and acute leukemia. Some of the classic drugs known to cause hematologic abnormalities have been replaced by newer drugs, including biologics, accompanied by their own syndromes and unintended side effects. Conclusions. Drugs can induce toxicities spanning many hematologic syndromes, mediated by a variety of mechanisms. Physicians need to be alert to the potential for iatrogenic drug-induced hematologic complications.
Collapse
|
244
|
Sud R, Khorana AA. Cancer-associated thrombosis: risk factors, candidate biomarkers and a risk model. Thromb Res 2009; 123 Suppl 4:S18-21. [PMID: 19303497 DOI: 10.1016/s0049-3848(09)70137-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer and its treatments are well-recognized risk factors for venous thrombo-embolism (VTE). Although solid tumors have historically been associated with VTE, more recent data suggest similar risk in patients with hematologic malignancies as well. The risk of VTE is not equal for all cancer patients or even in the same patient over time. In addition to well-known risk factors such as tumor site, stage, chemotherapy and comorbidities, candidate biomarkers have recently been identified including platelet and leukocyte counts, tissue factor and P-selectin. A validated risk model incorporating some of these risk factors and biomarkers has been shown to be predictive of VTE in cancer. VTE is associated with mortality, morbidity, potential delay in treatments for cancer and consumption of scarce health-care resources. Hence, reducing VTE with targeted prophylaxis could improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Rohit Sud
- James P. Wilmot Cancer Center, Department of Medicine, University of Rochester, NY, USA
| | | |
Collapse
|
245
|
Tzekova V, Mihaylov G, Elezovic I, Koytchev R. Therapeutic effects of epoetin zeta in the treatment of chemotherapy-induced anaemia. Curr Med Res Opin 2009; 25:1689-97. [PMID: 19505200 DOI: 10.1185/03007990903050876] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To perform an open, non-controlled, multiple-dose, international, multicentre, phase III study to evaluate epoetin zeta, a biosimilar epoetin referenced to epoetin alfa, for the treatment of chemotherapy-induced anaemia in patients with cancer. METHODS Safety, tolerability and efficacy of subcutaneously administered epoetin zeta were assessed in 216 patients with solid tumours or non-myeloid haematological malignancies receiving chemotherapy and at risk of transfusion. RESULTS A significant (p < 0.0001) increase in mean haemoglobin (Hb) level (1.8 g/dL) was observed between baseline and week 12 (intent-to-treat population); 176/216 (81.5%) patients achieved a response (increase in Hb > or = 1 g/dL or reticulocyte count > or = 40,000 cells/microL) by week 8. Over the treatment period, 231 treatment-emergent adverse events were experienced by 91 patients; 9/216 (4.2%) experienced a clinically significant thrombotic event within the first 12 weeks of epoetin zeta treatment, significantly lower than the assumed 18% baseline incidence (p < 0.0001) based on historical data from epoetin trials. No transfusion was necessary for 175/216 patients (81.0%) and quality of life improved over the study. No patients developed anti-erythropoietin antibodies. Sponsor trial no: CT-830-05-0009. CONCLUSION This study demonstrates that subcutaneously administered epoetin zeta is well-tolerated and has efficacy in the treatment of anaemia in patients with cancer receiving chemotherapy and at risk of transfusion.
Collapse
|
246
|
Treatment of myelodysplastic syndrome patients with erythropoietin with or without granulocyte colony-stimulating factor: results of a prospective randomized phase 3 trial by the Eastern Cooperative Oncology Group (E1996). Blood 2009; 114:2393-400. [PMID: 19564636 DOI: 10.1182/blood-2009-03-211797] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This phase 3 prospective randomized trial evaluated the efficacy and long-term safety of erythropoietin (EPO) with or without granulocyte colony-stimulating factor plus supportive care (SC; n = 53) versus SC alone (n = 57) for the treatment of anemic patients with lower-risk myelodysplastic syndromes. The response rates in the EPO versus SC alone arms were 36% versus 9.6%, respectively, at the initial treatment step, 47% in the EPO arm, including subsequent steps. Responding patients had significantly lower serum EPO levels (45% vs 5% responses for levels < 200 mU/mL vs > or = 200 mU/mL) and improvement in multiple quality-of-life domains. With prolonged follow-up (median, 5.8 years), no differences were found in overall survival of patients in the EPO versus SC arms (median, 3.1 vs 2.6 years) or in the incidence of transformation to acute myeloid leukemia (7.5% and 10.5% patients, respectively). Increased survival was demonstrated for erythroid responders versus nonresponders (median, 5.5 vs 2.3 years). Flow cytometric analysis showed that the percentage of P-glycoprotein(+) CD34(+) marrow blasts was positively correlated with longer overall survival. In comparison with SC alone, patients receiving EPO with or without granulocyte colony-stimulating factor plus SC had improved erythroid responses, similar survival, and incidence of acute myeloid leukemia transformation.
Collapse
|
247
|
Abstract
BACKGROUND The ability to maintain dialysis patients' hemoglobin (Hgb) within narrow targets remains a significant clinical problem. This study was designed to determine the variability in Hgb values for patients with chronic kidney disease (CKD) receiving or not receiving erythropoiesis-stimulating agents (ESAs) compared with patients on dialysis receiving ESAs. METHODS This cross-sectional review of anemia management in CKD and dialysis patients analyzed Hgb variability by patient-year, defined as the coefficient of variability calculated for individual patients. One hundred thirty-seven CKD patient-years and 350 dialysis patient-years were available for analysis. Hgb variability was defined as the coefficient of variability calculated as the individual patient's Hgb standard deviation divided by the patient's mean Hgb times 100. RESULTS The coefficient of variability in Hgb values were significantly less in patients with CKD not treated with ESAs than in patients with CKD treated with ESAs whether they were receiving dialysis (medians: 3.96 versus 8.53%, P < 0.05) or not receiving dialysis (medians: 3.96 versus 7.37%, P < 0.05). CONCLUSION CKD and hemodialysis patients receiving treatment with ESAs have significantly greater Hgb variability than patients with CKD not receiving ESAs. This finding suggests that the current practice pattern for the administration of exogenous ESAs is partly responsible for the observed Hgb variability.
Collapse
|
248
|
HEMATOLOGIC MALIGNANCIES. Cancer Invest 2009. [DOI: 10.1080/07357900701571858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
249
|
Ludwig H, Crawford J, Österborg A, Vansteenkiste J, Henry DH, Fleishman A, Bridges K, Glaspy JA. 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; 27:2838-47. [DOI: 10.1200/jco.2008.19.1130] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Although numerous clinical trials have demonstrated the efficacy and tolerability of erythropoiesis-stimulating agents (ESAs) in patients with chemotherapy-induced anemia (CIA), results of some recent trials and one meta-analysis have suggested that ESAs may negatively impact survival and/or disease control in patients with cancer. Methods To assess the benefits and risks of ESAs in CIA, we conducted a pooled analysis of individual patient-level data from all randomized, double-blind, placebo-controlled trials in 2,122 patients with CIA receiving darbepoetin alfa (DA; n = 1,200) or placebo (n = 912). Results DA did not increase mortality (hazard ratio = 0.97; 95% CI, 0.85 to 1.1) and had no effect on progression-free survival (hazard ratio = 0.93; 95% CI, 0.84 to 1.04) and disease progression (hazard ratio = 0.92; 95% CI, 0.82 to 1.03), but, as expected, increased the risk for thromboembolic events (hazard ratio = 1.57; 95% CI, 1.10 to 2.26). Overall and progression-free survival were not affected by baseline hemoglobin and seemed better in patients who achieved hemoglobin more than 12 or more than 13 g/dL. Transfusions and rates of hemoglobin increase (> 1 g/dL in 14 days; > 2 g/dL in 28 days) owing to transfusions were associated with an increased risk for death and disease progression in both treatment groups; in the absence of transfusions, rates of hemoglobin increase did not appear to increase the risk for adverse outcomes. Compared with placebo, DA significantly reduced the risk of receiving one or more transfusion. Conclusion There seemed to be no association between DA and risk of death or disease progression in this meta-analysis of individual patient data from DA studies conducted in CIA, the approved indication for ESAs in oncology.
Collapse
Affiliation(s)
- Heinz Ludwig
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| | - Jeffrey Crawford
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| | - Anders Österborg
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| | - Johan Vansteenkiste
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| | - David H. Henry
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| | - Alex Fleishman
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| | - Ken Bridges
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| | - John A. Glaspy
- From the Wilhelminenspital, Department of Medicine I, Center for Oncology and Hematology, Vienna, Austria; Duke University Medical Center, Durham, NC; Departments of Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden; Respiratory Oncology Unit, Department of Pulmonology, University Hospital Gasthuisberg, Leuven, Belgium; Pennsylvania Oncology Hematology Associates, Philadelphia, PA; Amgen Inc, Thousand Oaks; and University of California, Los Angeles, School of Medicine, Los
| |
Collapse
|
250
|
Wauters I, Vansteenkiste J. Darbepoetin alfa in the treatment of chemotherapy-induced anaemia. Expert Opin Biol Ther 2009; 9:221-30. [PMID: 19236252 DOI: 10.1517/14712590802652280] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Darbepoetin alfa is an erythropoiesis-stimulating agent (ESA), with a longer half-life than previous recombinant human erythropoietins. After its initial development for anaemia due to renal insufficiency, an extensive clinical trial program has defined its role in cancer patients. OBJECTIVE/METHODS Review of the initial registration studies, further development and recent progress, guidelines for use in clinical practice (EORTC, ASCO/ASH), and specific focus on recent safety concerns. RESULTS Darbepoetin alfa significantly decreases the number of red blood cell transfusions in patients with chemotherapy-induced anaemia, and has been shown to improve health-related quality of life in several studies. The prolonged half-life allows a prolonged dosing interval. Administration every three weeks, a suitable schedule to synchronise with day 1 of many chemotherapy regimens, is as efficient as the initially registered weekly administration. Recent data strongly suggest that the addition of intravenous iron improves haemoglobin response rates. The use of these agents in clinical practice has to be according to the guidelines. Recent safety data reported a negative effect on survival when ESAs were used to treat anaemia that was either not chemotherapy related, or when used to maintain high levels of haemoglobin and prevent anaemia. All of these studies were not in accordance with existing guidelines, while safety data from clinical trials using ESAs according to the guidelines remain reassuring. CONCLUSION Darbepoetin alfa has a well defined place in the treatment of chemotherapy-induced anaemia, and is safe when used in line with existing guidelines. Recent safety signals on cancer outcomes in studies not in accordance with these guidelines illustrate the need for further research into the complex interaction between anaemia and tumour hypoxia in cancer patients.
Collapse
Affiliation(s)
- Isabelle Wauters
- Catholic University, University Hospital Gasthuisberg, Respiratory Oncology Unit (Pulmonology) and Leuven Lung Cancer Group, Herestraat 49, B-3000 Leuven, Belgium
| | | |
Collapse
|