51
|
Caruso RA, Branca G, Fedele F, Irato E, Finocchiaro G, Parisi A, Ieni A. Mechanisms of coagulative necrosis in malignant epithelial tumors (Review). Oncol Lett 2014; 8:1397-1402. [PMID: 25202341 PMCID: PMC4156238 DOI: 10.3892/ol.2014.2345] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 07/01/2014] [Indexed: 01/10/2023] Open
Abstract
Histological tumor necrosis (TN) has been reported to indicate a poor prognosis for different human cancers. It is generally accepted that TN results from chronic ischemic injury due to rapid tumor growth. However, whether insufficient tumor vascularization and inadequate tumor cell oxygenation are the only factors causing TN remains controversial. Mitotic catastrophe is considered to occur as a result of dysregulated/failed mitosis, leading to cell death. We hypothesize that mitotic catastrophe, induced by hypoxic stress, may lead to the TN which is observed in high grade carcinomas. The current review describes the morphological features of TN in malignant epithelial tumors. In addition, evidence regarding the involvement of mitotic catastrophe in the induction of TN in human carcinomas is discussed.
Collapse
Affiliation(s)
- Rosario A Caruso
- Department of Human Pathology, University of Messina, Messina I-98125, Italy
| | - Giovanni Branca
- Department of Human Pathology, University of Messina, Messina I-98125, Italy
| | - Francesco Fedele
- Department of Human Pathology, University of Messina, Messina I-98125, Italy
| | - Eleonora Irato
- Department of Human Pathology, University of Messina, Messina I-98125, Italy
| | | | - Antonio Parisi
- Department of Surgical Science, University of Messina, Messina I-98125, Italy
| | - Antonio Ieni
- Department of Human Pathology, University of Messina, Messina I-98125, Italy
| |
Collapse
|
52
|
Zhou B, Damrauer JS, Bailey ST, Hadzic T, Jeong Y, Clark K, Fan C, Murphy L, Lee CY, Troester MA, Miller CR, Jin J, Darr D, Perou CM, Levine RL, Diehn M, Kim WY. Erythropoietin promotes breast tumorigenesis through tumor-initiating cell self-renewal. J Clin Invest 2014; 124:553-63. [PMID: 24435044 DOI: 10.1172/jci69804] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 10/24/2013] [Indexed: 12/30/2022] Open
Abstract
Erythropoietin (EPO) is a hormone that induces red blood cell production. In its recombinant form, EPO is the one of most prescribed drugs to treat anemia, including that arising in cancer patients. In randomized trials, EPO administration to cancer patients has been associated with decreased survival. Here, we investigated the impact of EPO modulation on tumorigenesis. Using genetically engineered mouse models of breast cancer, we found that EPO promoted tumorigenesis by activating JAK/STAT signaling in breast tumor-initiating cells (TICs) and promoted TIC self renewal. We determined that EPO was induced by hypoxia in breast cancer cell lines, but not in human mammary epithelial cells. Additionally, we demonstrated that high levels of endogenous EPO gene expression correlated with shortened relapse-free survival and that pharmacologic JAK2 inhibition was synergistic with chemotherapy for tumor growth inhibition in vivo. These data define an active role for endogenous EPO in breast cancer progression and breast TIC self-renewal and reveal a potential application of EPO pathway inhibition in breast cancer therapy.
Collapse
|
53
|
Wan S, Lai Y, Myers RE, Li B, Palazzo JP, Burkart AL, Chen G, Xing J, Yang H. Post-diagnosis hemoglobin change associates with overall survival of multiple malignancies - results from a 14-year hospital-based cohort of lung, breast, colorectal, and liver cancers. BMC Cancer 2013; 13:340. [PMID: 23841898 PMCID: PMC3710492 DOI: 10.1186/1471-2407-13-340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 07/05/2013] [Indexed: 12/04/2022] Open
Abstract
Background Anemia refers to low hemoglobin (Hb) level and is a risk factor of cancer patient survival. The National Comprehensive Cancer Network recently suggested that post-diagnosis Hb change, regardless of baseline Hb level, indicates the potential presence of anemia. However, there is no epidemiological study evaluating whether Hb change has direct prognostic values for cancer patients at the population level. Methods We identified 6675 patients with a diagnosis of primary lung, breast, colorectal, or liver cancer who visited the Kimmel Cancer Center at the Thomas Jefferson University from 1998 to 2011. All patients had at least two Hb measurements within the first six months after diagnosis. We analyzed the main, dose-dependent, and time-dependent effects of Hb changes on patient survival. Results Compared to patients with a low Hb change (|∆Hb|≤2.6), those having a |∆Hb|>2.6 exhibited a significantly shorter survival (hazard ratio=1.40, 95% confidence interval 1.31-1.50, P=4.5 × 10-22, Plog rank=1.6 × 10-39). This association remained significant across the four cancer types. Bootstrap resampling validated these findings 100% of the time with P<0.01 in all patients and in patients of individual cancers. The association exhibited an apparent U-shape dose-dependent pattern. Time-dependent modeling demonstrated that the effect of Hb change on the survival of the overall patient population persisted for approximately 4.5 years after diagnosis. Conclusion Post-diagnosis Hb change associates with the survival of multiple cancers and may have clinical values in tailoring anti-anemia treatments. Because Hb level is frequently measured during cancer treatment, Hb changes may be a potentially important variable in building cancer prognosis models.
Collapse
Affiliation(s)
- Shaogui Wan
- Division of Population Science, Department of Medical Oncology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Roberts EW, Deonarine A, Jones JO, Denton AE, Feig C, Lyons SK, Espeli M, Kraman M, McKenna B, Wells RJ, Zhao Q, Caballero OL, Larder R, Coll AP, O’Rahilly S, Brindle KM, Teichmann SA, Tuveson DA, Fearon DT. Depletion of stromal cells expressing fibroblast activation protein-α from skeletal muscle and bone marrow results in cachexia and anemia. J Exp Med 2013; 210:1137-51. [PMID: 23712428 PMCID: PMC3674708 DOI: 10.1084/jem.20122344] [Citation(s) in RCA: 324] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 04/29/2013] [Indexed: 12/15/2022] Open
Abstract
Fibroblast activation protein-α (FAP) identifies stromal cells of mesenchymal origin in human cancers and chronic inflammatory lesions. In mouse models of cancer, they have been shown to be immune suppressive, but studies of their occurrence and function in normal tissues have been limited. With a transgenic mouse line permitting the bioluminescent imaging of FAP(+) cells, we find that they reside in most tissues of the adult mouse. FAP(+) cells from three sites, skeletal muscle, adipose tissue, and pancreas, have highly similar transcriptomes, suggesting a shared lineage. FAP(+) cells of skeletal muscle are the major local source of follistatin, and in bone marrow they express Cxcl12 and KitL. Experimental ablation of these cells causes loss of muscle mass and a reduction of B-lymphopoiesis and erythropoiesis, revealing their essential functions in maintaining normal muscle mass and hematopoiesis, respectively. Remarkably, these cells are altered at these sites in transplantable and spontaneous mouse models of cancer-induced cachexia and anemia. Thus, the FAP(+) stromal cell may have roles in two adverse consequences of cancer: their acquisition by tumors may cause failure of immunosurveillance, and their alteration in normal tissues contributes to the paraneoplastic syndromes of cachexia and anemia.
Collapse
Affiliation(s)
- Edward W. Roberts
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Andrew Deonarine
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - James O. Jones
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Alice E. Denton
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Christine Feig
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Scott K. Lyons
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Marion Espeli
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Matthew Kraman
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Brendan McKenna
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Richard J.B. Wells
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Qi Zhao
- Ludwig Collaborative Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Otavia L. Caballero
- Ludwig Collaborative Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Rachel Larder
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Anthony P. Coll
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Stephen O’Rahilly
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Kevin M. Brindle
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - Sarah A. Teichmann
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| | - David A. Tuveson
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - Douglas T. Fearon
- Department of Medicine; Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council (MRC) Building; and Institute of Metabolic Sciences; Addenbrooke’s Hospital; Cancer Research UK Cambridge Institute, Li Ka Shing Centre; and MRC Laboratory of Molecular Biology; University of Cambridge, Cambridge CB2 2QH, England, UK
| |
Collapse
|
55
|
Nantel-Battista M, Vadeboncoeur S, Benohanian A. Selection of safe parameters for jet injection of botulinum toxin in palmar hyperhidrosis. Aesthet Surg J 2013; 33:295-7. [PMID: 23388650 DOI: 10.1177/1090820x12471675] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
56
|
Anees Ahmed RA, Ganvir SM, Hazarey VK. Relation of erythrocyte indices and serum iron level with clinical and histological progression of oral squamous cell carcinoma in central India. ACTA ACUST UNITED AC 2012; 5:65-71. [DOI: 10.1111/jicd.12021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/21/2012] [Indexed: 11/30/2022]
Affiliation(s)
- Riyaz Ahmed Anees Ahmed
- Department of Oral Pathology and Microbiology; Government Dental College and Hospital; Nagpur Maharashtra India
| | - Sindhu M. Ganvir
- Department of Oral Pathology and Microbiology; Government Dental College and Hospital; Nagpur Maharashtra India
| | - Vinay K. Hazarey
- Department of Oral Pathology and Microbiology; Government Dental College and Hospital; Nagpur Maharashtra India
| |
Collapse
|
57
|
Panneerselvam J, Park HK, Zhang J, Dudimah FD, Zhang P, Wang H, Fei P. FAVL impairment of the Fanconi anemia pathway promotes the development of human bladder cancer. Cell Cycle 2012; 11:2947-55. [PMID: 22828653 PMCID: PMC3419064 DOI: 10.4161/cc.21400] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Effectiveness of DNA cross-linking drugs in the treatment of bladder cancer suggests that bladder cancer cells may have harbored an insufficient cellular response to DNA cross-link damage, which will sensitize cells to DNA cross-linking agents. Cell sensitivity benefits from deficient DNA damage responses, which, on the other hand, can cause cancer. Many changed cellular signaling pathways are known to be involved in bladder tumorigenesis; however, DNA cross-link damage response pathway [Fanconi anemia (FA) pathway], whose alterations appear to be a plausible cause of the development of bladder cancer, remains an under-investigated area in bladder cancer research. In this study, we found FAVL (variant of FA protein L--FANCL) was elevated substantially in bladder cancer tissues examined. Ectopic expression of FAVL in bladder cancer cells as well as normal human cells confer an impaired FA pathway and hypersensitivity to Mitomycin C, similar to those found in FA cells, indicating that FAVL elevation may possess the same tumor promotion potential as an impaired FA pathway harbored in FA cells. Indeed, a higher level of FAVL expression can promote the growth of bladder cancer cells in vitro and in vivo, which, at least partly, results from FAVL perturbation of FANCL expression, an essential factor for the activation of the FA pathway. Moreover, a higher level of FAVL expression was found to be associated with chromosomal instability and the invasiveness of bladder cancer cells. Collectively, FAVL elevation can increase the tumorigenic potential of bladder cancer cells, including the invasive potential that confers the development of advanced bladder cancer. These results enhance our understanding the pathogenesis of human bladder cancer, holding a promise to develop additional effective tools to fight human bladder cancer.
Collapse
Affiliation(s)
| | - Hwan Ki Park
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester, MN USA
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester, MN USA
| | | | - Piyan Zhang
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester, MN USA
| | - Hong Wang
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester, MN USA
| | - Peiwen Fei
- University of Hawaii Cancer Center; University of Hawaii; Honolulu, HI USA
| |
Collapse
|
58
|
Fokas E, McKenna WG, Muschel RJ. The impact of tumor microenvironment on cancer treatment and its modulation by direct and indirect antivascular strategies. Cancer Metastasis Rev 2012; 31:823-42. [DOI: 10.1007/s10555-012-9394-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
59
|
Hallermann C, Niermann C, Fischer RJ, Schulze HJ. Erythrocyte sedimentation rate as an independent prognostic factor in mycosis fungoides. Br J Dermatol 2012; 166:873-4. [DOI: 10.1111/j.1365-2133.2011.10729.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
60
|
Richards CH, Roxburgh CSD, Anderson JH, McKee RF, Foulis AK, Horgan PG, McMillan DC. Prognostic value of tumour necrosis and host inflammatory responses in colorectal cancer. Br J Surg 2011; 99:287-94. [PMID: 22086662 DOI: 10.1002/bjs.7755] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2011] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tumour necrosis is a marker of poor prognosis in some tumours but the mechanism is unclear. This study examined the prognostic value of tumour necrosis and host inflammatory responses in colorectal cancer. METHODS This was a retrospective study of patients undergoing potentially curative resection of colorectal cancer at a single surgical institution over a 10-year period. Patients who underwent preoperative radiotherapy were excluded. The systemic and local inflammatory responses were assessed using the modified Glasgow Prognostic Score and Klintrup-Makinen criteria respectively. Original tumour sections were retrieved and necrosis graded as absent, focal, moderate or extensive. Associations between necrosis and clinicopathological variables were examined, and multivariable survival analyses carried out. RESULTS A total of 343 patients were included between 1997 and 2007. Tumour necrosis was graded as absent in 32 (9·3 per cent), focal in 166 (48·4 per cent), moderate in 101 (29·4 per cent) and extensive in 44 (12·8 per cent). There were significant associations between tumour necrosis and anaemia (P = 0·022), white cell count (P = 0·006), systemic inflammatory response (P < 0·001), local inflammatory cell infiltrate (P = 0·004), tumour node metastasis (TNM) stage (P = 0·015) and Petersen Index (P = 0·003). On univariable survival analysis, tumour necrosis was associated with cancer-specific survival (P < 0·001). On multivariable survival analysis, age (hazard ratio (HR) 1·29, 95 per cent confidence interval 1·00 to 1·66), systemic inflammatory response (HR 1·74, 1·27 to 2·39), low-grade local inflammatory cell infiltrate (HR 2·65, 1·52 to 4·63), TNM stage (HR 1·55, 1·02 to 2·35) and high-risk Petersen Index (HR 3·50, 2·21 to 5·55) were associated with reduced cancer-specific survival. CONCLUSION The impact of tumour necrosis on colorectal cancer survival may be due to close associations with the host systemic and local inflammatory responses.
Collapse
Affiliation(s)
- C H Richards
- University Department of Surgery, Glasgow Royal Infirmary, Glasgow, UK.
| | | | | | | | | | | | | |
Collapse
|
61
|
Chateauvieux S, Grigorakaki C, Morceau F, Dicato M, Diederich M. Erythropoietin, erythropoiesis and beyond. Biochem Pharmacol 2011; 82:1291-303. [DOI: 10.1016/j.bcp.2011.06.045] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 06/28/2011] [Accepted: 06/29/2011] [Indexed: 12/21/2022]
|
62
|
Baker JM, De Lisio M, Parise G. Endurance exercise training promotes medullary hematopoiesis. FASEB J 2011; 25:4348-57. [PMID: 21868472 DOI: 10.1096/fj.11-189043] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Endurance exercise is a poorly defined yet powerful mediator of hematopoiesis. The purpose of this study was to directly investigate the effects of endurance exercise training on hematopoiesis and to identify potential mechanisms responsible for any observed changes. Four-week-old male C57Bl/6 mice were trained on a treadmill at progressive speeds over a 10-wk period. Tissues were harvested 2 d following the final training session. Flow cytometry, the cobblestone area-forming cell assay, and the methycellulose colony-forming unit assay were used to assess medullary and mobilized hematopoietic stem and progenitor cells. Quantitative real-time PCR and Western blots were used to measure hematopoietic cytokine production. Histochemistry was also used to assess adaptations to exercise in the bone marrow niche. Depending on the cell type, endurance training increased medullary and mobilized hematopoietic stem and progenitor cell content from 50 to 800%. Training also reduced marrow cavity fat by 78%. Skeletal muscle hematopoietic cytokine expression was also increased at least 60% by training. Sedentary mice served as controls for the above experiments. In conclusion, endurance exercise training greatly promotes hematopoiesis and does so through improvements in medullary niche architecture as well as increased skeletal muscle hematopoietic cytokine production.
Collapse
Affiliation(s)
- J M Baker
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | | | | |
Collapse
|
63
|
Talbot AL, Bullock GC, Delehanty LL, Sattler M, Zhao ZJ, Goldfarb AN. Aconitase regulation of erythropoiesis correlates with a novel licensing function in erythropoietin-induced ERK signaling. PLoS One 2011; 6:e23850. [PMID: 21887333 PMCID: PMC3161794 DOI: 10.1371/journal.pone.0023850] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 07/26/2011] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Erythroid development requires the action of erythropoietin (EPO) on committed progenitors to match red cell output to demand. In this process, iron acts as a critical cofactor, with iron deficiency blunting EPO-responsiveness of erythroid progenitors. Aconitase enzymes have recently been identified as possible signal integration elements that couple erythropoiesis with iron availability. In the current study, a regulatory role for aconitase during erythropoiesis was ascertained using a direct inhibitory strategy. METHODOLOGY/PRINCIPAL FINDINGS In C57BL/6 mice, infusion of an aconitase active-site inhibitor caused a hypoplastic anemia and suppressed responsiveness to hemolytic challenge. In a murine model of polycythemia vera, aconitase inhibition rapidly normalized red cell counts, but did not perturb other lineages. In primary erythroid progenitor cultures, aconitase inhibition impaired proliferation and maturation but had no effect on viability or ATP levels. This inhibition correlated with a blockade in EPO signal transmission specifically via ERK, with preservation of JAK2-STAT5 and Akt activation. Correspondingly, a physical interaction between ERK and mitochondrial aconitase was identified and found to be sensitive to aconitase inhibition. CONCLUSIONS/SIGNIFICANCE Direct aconitase inhibition interferes with erythropoiesis in vivo and in vitro, confirming a lineage-selective regulatory role involving its enzymatic activity. This inhibition spares metabolic function but impedes EPO-induced ERK signaling and disturbs a newly identified ERK-aconitase physical interaction. We propose a model in which aconitase functions as a licensing factor in ERK-dependent proliferation and differentiation, thereby providing a regulatory input for iron in EPO-dependent erythropoiesis. Directly targeting aconitase may provide an alternative to phlebotomy in the treatment of polycythemia vera.
Collapse
Affiliation(s)
- Anne-Laure Talbot
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Grant C. Bullock
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Lorrie L. Delehanty
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Adam N. Goldfarb
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
64
|
Tumor necrosis factor alpha-mediated inhibition of erythropoiesis involves GATA-1/GATA-2 balance impairment and PU.1 over-expression. Biochem Pharmacol 2011; 82:156-66. [PMID: 21501595 DOI: 10.1016/j.bcp.2011.03.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 03/27/2011] [Accepted: 03/31/2011] [Indexed: 01/06/2023]
|
65
|
Kim JH, Lee JM, Ryu KS, Lee YS, Park YG, Hur SY, Lee KH, Lee SH. The prognostic impact of duration of anemia during chemotherapy in advanced epithelial ovarian cancer. Oncologist 2011; 16:1154-61. [PMID: 21705663 DOI: 10.1634/theoncologist.2010-0236] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To propose a measure of anemia to be used as a prognostic factor for progression-free survival and overall survival in advanced epithelial ovarian cancer patients. PATIENTS AND METHODS Seventy-six patients with International Federation of Gynecology and Obstetrics stage III and stage IV epithelial ovarian cancer who had received at least six courses of platinum- and taxane-based systemic chemotherapy and achieved clinical or pathologic complete response were included. A novel prognostic factor based on the duration of anemia was proposed and the impact of anemia on progression-free and overall survival times was analyzed by a log-rank test and a Cox proportional hazards model. RESULTS We introduce a binary variable, Hb1020, that takes a value of 1 if the duration of a hemoglobin (Hb) level <10 g/dL is ≥20% of the total duration of chemotherapy. We propose Hb1020 as a potential prognostic factor for epithelial ovarian cancer. The 5-year progression-free survival rates were 48.4% in the Hb1020 = 0 group (duration of Hb <10 g/dL <20% of total duration) and 17.7% in the Hb1020 = 1 group (p = .026). The 5-year overall survival rates were 64.6% and 45.0%, respectively (p = .015). CONCLUSIONS Hb1020, based on the duration of anemia, is a potential prognostic factor for epithelial ovarian cancer. Using Hb1020, we will be able to administer highly optimized treatment for anemia to improve patient survival. Further independent studies are needed to confirm its prognostic role.
Collapse
Affiliation(s)
- Jin Hwi Kim
- Department of Obstetrics Gynecology, Catholic University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
66
|
[Red blood cells transfusions in oncological patients treated with radio- and chemotherapy]. VOJNOSANIT PREGL 2011; 68:28-34. [PMID: 21425615 DOI: 10.2298/vsp1101028a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIM Anemia is one of the most frequent hematology disorders in patients with malignant diseases. It has a great influence on reduction of the quality of life, so it requires early diagnosis and an adequate treatment. The aim of this study was to present and analyze the treatment of anemia using red blood cell transfusions in patients with malignancies, to analyze adequate use of red blood cell transfusions according to hemoglobin concentration, and also the influence of the treatment of malignant disease on the level of anemia and use of red blood cells transfusion. METHODS This retrospective analysis included the data on the use of red blood cells in Oncological Clinic of Clinical Center Nis in a period from the 1st January 2008 to the 31st December 2008. RESULTS None of the patients received the whole blood. In this period, 735 patients received 1006 units of red blood cells (red blood cell concentrate, resuspended, washed, filtered). An average use of red blood cell transfusion was 1.37 unit per oncological patient who received transfusion. The use of red blood cell units was adequate (87.60% of patients received transfusion of red cells when Hgb < 80 g/L). During radio- and chemotherapy we noticed a decrease of hematological parameter values. The patients of the experimental group were dependant on red blood cells transfusion. Statistically, a significant decrease of hemoglobin level was observed in patients treated only with radiotherapy who are the greatest consumers of red blood cells. Two patients were registered who more likely to have febrile nonhemolytic transfusion reactions. Posttransfusion alloimmunization occurred in 0.68% of the patients. CONCLUSION The use of red blood cells in oncological patients is in compliance with the up to date tendencies and recommendations published in clinical guidelines. For the purpose of efficient transfusion support in patients with malignant diseases, we have to follow the newest knowledge and attitudes about clinical use of blood products.
Collapse
|
67
|
Sebastiani G, Pantopoulos K. Disorders associated with systemic or local iron overload: from pathophysiology to clinical practice. Metallomics 2011; 3:971-86. [DOI: 10.1039/c1mt00082a] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
68
|
Steensma DP, Sloan JA, Dakhil SR, Dalton R, Kahanic SP, Prager DJ, Stella PJ, Rowland KM, Novotny PJ, Loprinzi CL. Phase III, randomized study of the effects of parenteral iron, oral iron, or no iron supplementation on the erythropoietic response to darbepoetin alfa for patients with chemotherapy-associated anemia. J Clin Oncol 2011; 29:97-105. [PMID: 21098317 PMCID: PMC3055863 DOI: 10.1200/jco.2010.30.3644] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 09/23/2010] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Functional iron deficiency may impair response to erythropoiesis-stimulating agents (ESAs) in iron-replete patients with chemotherapy-associated anemia (CAA). This study evaluated whether coadministration of parenteral iron improves ESA efficacy in patients with CAA. PATIENTS AND METHODS This prospective, multicenter, randomized trial enrolled 502 patients with hemoglobin (Hb) less than 11 g/dL who were undergoing chemotherapy for nonmyeloid malignancies. All patients received darbepoetin alfa once every 3 weeks and were randomly assigned to receive either ferric gluconate 187.5 mg intravenously (IV) every 3 weeks, oral daily ferrous sulfate 325 mg, or oral placebo for 16 weeks. RESULTS There was no difference in the erythropoietic response rate (ie, proportion of patients achieving Hb ≥ 12 g/dL or Hb increase ≥ 2 g/dL from baseline): 69.5% (95% CI, 61.9% to 76.5%) of IV iron-treated patients achieved an erythropoietic response compared with 66.9% (95% CI, 59.1% to 74.0%) who received oral iron and 65.0% (95% CI, 57.2% to 72.3%) who received oral placebo (P = .75). There were also no differences in the proportion of patients requiring red cell transfusions, changes in quality of life, or the dose of darbepoetin administered. Adverse events (AEs) tended to be more common in the IV iron arm: grade 3 or higher AEs occurred in 54% (95% CI, 46% to 61%) of patients receiving IV iron compared with 44% (95% CI, 36% to 52%) who received oral iron and 46% (95% CI, 38% to 54%) who received oral placebo (P = .16). CONCLUSION In patients with CAA, addition of IV ferric gluconate to darbepoetin failed to provide additional benefit compared with oral iron or oral placebo.
Collapse
Affiliation(s)
- David P Steensma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney St, Suite D1B30, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Efficacy and Safety of Oral Lactoferrin Supplementation in Combination with rHuEPO‐β for the Treatment of Anemia in Advanced Cancer Patients Undergoing Chemotherapy: Open‐Label, Randomized Controlled Study. Oncologist 2010; 15:894-902. [PMID: 20647390 DOI: 10.1634/theoncologist.2010-0020] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
70
|
Kodippili GC, Spector J, Kang GE, Liu H, Wickrema A, Ritchie K, Low PS. Analysis of the kinetics of band 3 diffusion in human erythroblasts during assembly of the erythrocyte membrane skeleton. Br J Haematol 2010; 150:592-600. [PMID: 20553270 DOI: 10.1111/j.1365-2141.2010.08268.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During definitive erythropoiesis, erythroid precursors undergo differentiation through multiple nucleated states to an enucleated reticulocyte, which loses its residual RNA/organelles to become a mature erythrocyte. Over the course of these transformations, continuous changes in membrane proteins occur, including shifts in protein abundance, rates of expression, isoform prominence, states of phosphorylation, and stability. In an effort to understand when assembly of membrane proteins into an architecture characteristic of the mature erythrocyte occurs, we quantitated the lateral diffusion of the most abundant membrane protein, band 3 (AE1), during each stage of erythropoiesis using single particle tracking. Analysis of the lateral trajectories of individual band 3 molecules revealed a gradual reduction in mobility of the anion transporter as erythroblasts differentiated. Evidence for this progressive immobilization included a gradual decline in diffusion coefficients as determined at a video acquisition rate of 120 frames/s and a decrease in the percentage of compartment sizes >100 nm. Because complete acquisition of the properties of band 3 seen in mature erythrocytes is not observed until circulating erythrocytes are formed, we suggest that membrane maturation involves a gradual and cooperative assembly process that is not triggered by the synthesis of any single protein.
Collapse
|
71
|
Nurgalieva Z, Liu CC, Du XL. Chemotherapy use and risk of bone marrow suppression in a large population-based cohort of older women with breast and ovarian cancer. Med Oncol 2010; 28:716-25. [PMID: 20361359 DOI: 10.1007/s12032-010-9512-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 03/19/2010] [Indexed: 11/29/2022]
Abstract
We studied 65,521 women with breast cancer and 7,420 women with ovarian cancer aged ≥ 65 identified from the 16 areas of the Surveillance, Epidemiology and End Results program linked with Medicare data during 1991-2002. Bone marrow toxicity associated with chemotherapy was defined using diagnosis codes from Medicare inpatient, outpatient and physician claims. The time to event Cox regression was utilized to estimate the risk of bone marrow toxicity. Use of anthracyclines, taxanes or platinums was associated with increased risks of short- (≤3 months) and long-term (>3 months) anemia and neutropenia in patients with breast cancer. Alkylating agents or antimetabolites were additional significant predictors of anemia in women with ovarian cancer. Patients who received chemotherapy (irrespective of regimens) were twice (breast cancer) or three times (ovarian cancer) as likely to develop thrombocytopenia compared to those not receiving chemotherapy. Among women with breast cancer, patients receiving cyclophosphamide, methotrexate and fluorouracil regimens (hazard ratio=19.0, 95% CI=11.2-32.5), platinum/taxane therapy (21.9, 11.9-40.4) or the cyclophosphamide, adriamycin and fluorouracil regimen (32.5, 19.6-53.9) were strongly associated with risk of aplastic anemia. There was a dose-response relationship between the use of taxane or platinum and the risk of bone marrow suppression, whereas the increased risk of bone marrow toxicity was consistently higher in those with use of alkylating agents or anthracycline-based regimens irrespective of the increasing number of cycles received. In conclusion, there was an association between chemotherapy use and clinical manifestations of bone marrow toxicities in a population-based setting.
Collapse
Affiliation(s)
- Zhannat Nurgalieva
- Division of Epidemiology and Disease Control, University of Texas School of Public Health, 1200 Herman Pressler Drive, RAS-E631, Houston, TX, USA
| | | | | |
Collapse
|
72
|
Pelekanou V, Notas G, Sanidas E, Tsapis A, Castanas E, Kampa M. Testosterone membrane-initiated action in breast cancer cells: Interaction with the androgen signaling pathway and EPOR. Mol Oncol 2010; 4:135-49. [PMID: 20189893 PMCID: PMC5527892 DOI: 10.1016/j.molonc.2010.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 01/21/2010] [Accepted: 01/22/2010] [Indexed: 12/29/2022] Open
Abstract
Membrane-initiated androgen actions have now been acknowledged, even though a specific binding site has not been biochemically characterized yet. Recent data indicate that testosterone-BSA, a non-permeable testosterone analog, can exert specific actions in breast cancer cell lines, including proper transcriptional effects, independent of the intracellular androgen sites. In the present work we explore the effects of testosterone-BSA in two specifically modified pathways, revealed by early trascriptome analysis, namely the non-genotropic androgen signaling and the HIF1alpha pathway. We provide evidence that p38 MAPK and PI3K/Akt/NFkappaB and/or Rho/Actin pathways are directly involved in testosterone-induced apoptosis, while the JNK/c-JUN pathway is involved in membrane site-initiated transcription. Furthermore we show that membrane-acting androgens modify the transcription of the erythropoietin receptor (EPOR), leading to erythropoietin-initiated actions. Interestingly, association of recombinant human erythropoietin (rHuEPO) together with testosterone-BSA protects cells from apoptosis, through discrete signaling events. The effect of testosterone-BSA is exerted through the classical erythropoietin promoter, while rHuEPO decreases the transcription of EPOR acting on a newly identified regulatory/promoter region, upstream of its known promoter. These results suggest a new interaction of membrane-acting androgen with EPOR and should be taken into account in the pharmaceutical manipulations of breast cancer patients.
Collapse
Affiliation(s)
- Vassiliki Pelekanou
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| | - Elias Sanidas
- Department of Surgical Oncology, University of Crete, School of Medicine, Heraklion GR-71003, Greece
| | - Andreas Tsapis
- INSERM, U976, Paris, F-75010 France
- INSERM, Univ Paris-Diderot, Paris, F-75013 France
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, University of Crete, School of Medicine, P.O. Box 2208, Heraklion GR-71003, Greece
| |
Collapse
|
73
|
Abstract
Anaemia is a frequent complication of prostate cancer and of its treatments. In Europe prostate cancer accounts for the 10.8% of all malignant neoplasms. Iatrogenic hypogonadism and age-related physiologic changes along with nutritional deficits contribute to increase prevalence of prostate cancer related anaemia. The reason of the present review is to provide clinicians with all aspects of a frequent and multifactorial co-morbidity, whose effects are often underestimated. Erythropoiesis pathology and causes of anaemia in prostate cancer are reviewed. Critical issues of clinical management of anaemia in prostate cancer are discussed.
Collapse
Affiliation(s)
- Giuseppe Colloca
- Division of Medical Oncology, ASL-1 Imperiese, Ospedale Civile, Sanremo, Italy
| | | | | | | |
Collapse
|
74
|
Pro-inflammatory cytokine-mediated anemia: regarding molecular mechanisms of erythropoiesis. Mediators Inflamm 2010; 2009:405016. [PMID: 20204172 PMCID: PMC2830572 DOI: 10.1155/2009/405016] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Accepted: 12/17/2009] [Indexed: 12/26/2022] Open
Abstract
Anemia of cancer and chronic inflammatory diseases is a frequent complication affecting quality of life. For cancer patients it represents a particularly bad prognostic. Low level of erythropoietin is considered as one of the causes of anemia in these pathologies. The deficiency in erythropoietin production results from pro-inflammatory cytokines effect. However, few data is available concerning molecular mechanisms involved in cytokine-mediated anemia. Some recent publications have demonstrated the direct effect of pro-inflammatory cytokines on cell differentiation towards erythroid pathway, without erythropoietin defect. This suggested that pro-inflammatory cytokine-mediated signaling pathways affect erythropoietin activity. They could interfere with erythropoietin-mediated signaling pathways, inducing early apoptosis and perturbing the expression and regulation of specific transcription factors involved in the control of erythroid differentiation. In this review we summarize the effect of tumor necrosis factor (TNF)α, TNF-related apoptosis-inducing ligand (TRAIL), and interferon (IFN)-γ on erythropoiesis with a particular interest for molecular feature.
Collapse
|
75
|
Chang J, Wen B, Kazanzides P, Zanzonico P, Finn RD, Fichtinger G, Ling CC. A robotic system for 18F-FMISO PET-guided intratumoral pO2 measurements. Med Phys 2010; 36:5301-9. [PMID: 19994538 DOI: 10.1118/1.3239491] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An image-guided robotic system was used to measure the oxygen tension (pO2) in rodent tumor xenografts using interstitial probes guided by tumor hypoxia PET images. Rats with approximately 1 cm diameter tumors were anesthetized and immobilized in a custom-fabricated whole-body mold. Imaging was performed using a dedicated small-animal PET scanner (R4 or Focus 120 microPET) approximately 2 h after the injection of the hypoxia tracer 18F-fluoromisonidazole (18F-FMISO). The coordinate systems of the robot and PET were registered based on fiducial markers in the rodent bed visible on the PET images. Guided by the 3D microPET image set, measurements were performed at various locations in the tumor and compared to the corresponding 18F-FMISO image intensity at the respective measurement points. Experiments were performed on four tumor-bearing rats with 4 (86), 3 (80), 7 (162), and 8 (235) measurement tracks (points) for each experiment. The 18F-FMISO image intensities were inversely correlated with the measured pO2, with a Pearson coefficient ranging from -0.14 to -0.97 for the 22 measurement tracks. The cumulative scatterplots of pO2 versus image intensity yielded a hyperbolic relationship, with correlation coefficients of 0.52, 0.48, 0.64, and 0.73, respectively, for the four tumors. In conclusion, PET image-guided pO2 measurement is feasible with this robot system and, more generally, this system will permit point-by-point comparison of physiological probe measurements and image voxel values as a means of validating molecularly targeted radiotracers. Although the overall data fitting suggested that 18F-FMISO may be an effective hypoxia marker, the use of static 18F-FMISO PET postinjection scans to guide radiotherapy might be problematic due to the observed high variation in some individual data pairs from the fitted curve, indicating potential temporal fluctuation of oxygen tension in individual voxels or possible suboptimal imaging time postadministration of hypoxia-related trapping of 18F-FMISO.
Collapse
Affiliation(s)
- Jenghwa Chang
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA.
| | | | | | | | | | | | | |
Collapse
|
76
|
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
|
77
|
Edgren G, Bagnardi V, Bellocco R, Hjalgrim H, Rostgaard K, Melbye M, Reilly M, Adami HO, Hall P, Nyrén O. Pattern of declining hemoglobin concentration before cancer diagnosis. Int J Cancer 2009; 127:1429-36. [DOI: 10.1002/ijc.25122] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
78
|
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. 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: 76] [Impact Index Per Article: 4.8] [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
|
79
|
|
80
|
Inthal A, Krapf G, Beck D, Joas R, Kauer MO, Orel L, Fuka G, Mann G, Panzer-Grümayer ER. Role of the erythropoietin receptor in ETV6/RUNX1-positive acute lymphoblastic leukemia. Clin Cancer Res 2009; 14:7196-204. [PMID: 19010836 DOI: 10.1158/1078-0432.ccr-07-5051] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We explored the mechanisms leading to the distinct overexpression of EPOR as well as the effects of EPO signaling on ETV6/RUNX1-positive acute lymphoblastic leukemias. EXPERIMENTAL DESIGN ETV6/RUNX1-expressing model cell lines and leukemic cells were used for real-time PCR of EPOR expression. Proliferation, viability, and apoptosis were analyzed on cells exposed to EPO, prednisone, or inhibitors of EPOR pathways by [3H]thymidine incorporation, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and Annexin V/propidium iodide staining. Western blot analysis was done to detect activation of signaling proteins. Serum EPO levels and sequences of the EPOR (n = 53) as well as hemoglobin levels were taken from children with acute lymphoblastic leukemia enrolled in Austrian protocols. RESULTS We show here that ectopic expression of ETV6/RUNX1 induced EPOR up-regulation. Anemia, however, did not appear to influence EPOR expression on leukemic cells, although children with ETV6/RUNX1-positive leukemias had a lower median hemoglobin than controls. Exposure to EPO increased proliferation and survival of ETV6/RUNX1-positive leukemias in vitro, whereas blocking its binding site did not alter cell survival. The latter was not caused by activating mutations in the EPOR but might be triggered by constitutive activation of phosphatidylinositol 3-kinase/Akt, the major signaling pathway of EPOR in these cells. Moreover, prednisone-induced apoptosis was attenuated in the presence of EPO in this genetic subgroup. CONCLUSIONS Our data suggest that ETV6/RUNX1 leads to EPOR up-regulation and that activation by EPO might be of relevance to the biology of this leukemia subtype. Further studies are, however, needed to assess the clinical implications of its apoptosis-modulating properties.
Collapse
Affiliation(s)
- Andrea Inthal
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Albuquerque EX, Pereira EFR, Alkondon M, Rogers SW. Mammalian nicotinic acetylcholine receptors: from structure to function. Physiol Rev 2009; 89:73-120. [PMID: 19126755 PMCID: PMC2713585 DOI: 10.1152/physrev.00015.2008] [Citation(s) in RCA: 1263] [Impact Index Per Article: 78.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The classical studies of nicotine by Langley at the turn of the 20th century introduced the concept of a "receptive substance," from which the idea of a "receptor" came to light. Subsequent studies aided by the Torpedo electric organ, a rich source of muscle-type nicotinic receptors (nAChRs), and the discovery of alpha-bungarotoxin, a snake toxin that binds pseudo-irreversibly to the muscle nAChR, resulted in the muscle nAChR being the best characterized ligand-gated ion channel hitherto. With the advancement of functional and genetic studies in the late 1980s, the existence of nAChRs in the mammalian brain was confirmed and the realization that the numerous nAChR subtypes contribute to the psychoactive properties of nicotine and other drugs of abuse and to the neuropathology of various diseases, including Alzheimer's, Parkinson's, and schizophrenia, has since emerged. This review provides a comprehensive overview of these findings and the more recent revelations of the impact that the rich diversity in function and expression of this receptor family has on neuronal and nonneuronal cells throughout the body. Despite these numerous developments, our understanding of the contributions of specific neuronal nAChR subtypes to the many facets of physiology throughout the body remains in its infancy.
Collapse
Affiliation(s)
- Edson X Albuquerque
- Department of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
82
|
|
83
|
Bohlius J, Trelle S, Weingart O, Schwarzer G, Brillant C, Clarke MJ, Djulbegovic B, Piper M, Rades D, Seidenfeld J, Somerfield M, Steensma D, Schumacher M, Engert A, Egger M. Erythropoietin or Darbepoetin for patients with cancer - meta-analysis based on individual patient data. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2008. [DOI: 10.1002/14651858.cd007303] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
84
|
Strauss HG, Haensgen G, Dunst J, Hayward CRW, Burger HU, Scherhag A, Koelbl H. Effects of anemia correction with epoetin beta in patients receiving radiochemotherapy for advanced cervical cancer. Int J Gynecol Cancer 2008; 18:515-24. [PMID: 17645506 DOI: 10.1111/j.1525-1438.2007.01032.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Patients with cervical cancer frequently suffer from anemia. This two-stage, adaptive-design study investigated the effect of anemia correction with epoetin beta on treatment outcomes. Patients with stage IIB–IVA cervical cancer received radiochemotherapy (RCT) and were randomized to epoetin 150 IU/kg three times weekly (n = 34) or standard care (control; n = 40) for up to 12 weeks. Primary end point for stage 1 aimed to establish a correlation between anemia correction and treatment failure (no complete response or relapsing within 6 months after RCT initiation) as a proof of concept before moving into stage 2. Secondary end points included progression/relapse-free survival, overall survival, response to RCT, hemoglobin (Hb) response, and safety. Median baseline Hb was 11.4 and 11.6 g/dL in epoetin and control groups, respectively. At treatment end point, median Hb increased by 1.3 g/dL with epoetin, but decreased by 0.7 g/dL in the control group (P < 0.0001). No significant correlation between Hb increase and treatment failure was demonstrated. There were no significant differences between epoetin and control groups in progression/relapse-free survival (29.4% vs 32.5% patients with events; P = 0.96), overall survival (23.5% vs 12.5% patients with events; P = 0.22) or overall complete response (53% vs 58%; P = 0.86). Adverse events were well matched between groups. This study shows that epoetin beta rapidly, effectively, and safely increases Hb levels in patients with cervical cancer receiving RCT. No positive correlation of Hb increase and improvement in clinical outcomes could be demonstrated.
Collapse
Affiliation(s)
- H-G Strauss
- Department of Gynecology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | | | | | | | | | | | | |
Collapse
|
85
|
Guan X, Chen L. Role of erythropoietin in cancer-related anaemia: a double-edged sword? J Int Med Res 2008; 36:1-8. [PMID: 18230261 DOI: 10.1177/147323000803600101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Anaemia often occurs in cancer patients and its origin is multifactorial, resulting from either bone marrow infiltration of cancer cells or cytotoxic effects produced by chemotherapy and radiotherapy. Anaemia impacts significantly on quality of life and appears markedly to limit disease control. Erythropoietin stimulates erythrocyte formation and the human recombinant form is useful in treating anaemia in cancer patients. Over the past decade erythropoietin has been associated with amelioration of anaemia and reduced need for blood transfusions. Nevertheless, several pre-clinical and clinical trials, employing relatively high doses of erythropoietin, have been halted recently following increased mortality and morbidity, primarily due to thrombotic events and possible tumour growth stimulation. It is, therefore, too early to know whether erythropoietin is useful in controlling morbidity and mortality in cancer-related anaemia. The risk-benefit of erythropoietic agents should be studied in carefully controlled trials. This review discusses prevalent issues and addresses key questions concerning the use of erythropoietic agents for the treatment of cancer-related anaemia.
Collapse
Affiliation(s)
- X Guan
- Department of Oncology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | | |
Collapse
|
86
|
Association of abnormal preoperative laboratory values with survival after radical nephrectomy for clinically confined clear cell renal cell carcinoma. Urology 2008; 71:278-82. [PMID: 18308103 DOI: 10.1016/j.urology.2007.08.048] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2007] [Revised: 07/15/2007] [Accepted: 08/24/2007] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To determine whether preoperative laboratory values are independently associated with death from clinically confined clear cell renal cell carcinoma (RCC) after radical nephrectomy. METHODS We identified 1707 patients with clinically confined (pNx/pN0, pM0), unilateral, sporadic clear cell RCC treated with radical nephrectomy between 1970 and 2002. Associations of abnormal preoperative laboratory values including hypercalcemia, anemia, elevated erythrocyte sedimentation rate (ESR), and elevated alkaline phosphatase with death from RCC were evaluated using Cox proportional hazards regression models, both univariately and multivariately by adjusting for known prognostic features of the 2002 primary tumor classification, tumor size, nuclear grade, and coagulative tumor necrosis. RESULTS At last follow-up, 1009 patients had died, including 425 who died from RCC at a median of 3.0 years after surgery (range, 0 to 26 years). Even after adjusting for known prognostic features, 9% of patients with preoperative hypercalcemia exhibited significantly increased likelihood of dying from RCC compared with patients with normal or lower levels of serum calcium (relative ration [RR] 1.64; P = 0.002). Similarly, preoperative anemia (35% of patients; RR 1.27; P = 0.026) and elevated ESR (44% of patients; RR 1.66; P = 0.003) portended an increased risk of death from RCC even after multivariate adjustment. CONCLUSIONS Abnormal preoperative laboratory values including hypercalcemia, anemia, and elevated ESR are independently associated with increased risk of cancer-specific death from clinically confined clear cell RCC. Consideration of these variables in future models may improve prognostic accuracy. We believe these factors should be routinely assessed and included in prospective studies of outcome in RCC patients.
Collapse
|
87
|
Abstract
Many recombinant growth factors have failed in clinical trials due to off-target effects. We describe a method for circumventing off-target effects that involves equipping cells with a conditionally active signaling protein that can be specifically activated by an exogenously administered synthetic ligand. We believe that this approach will have many applications in gene and cell therapy.
Collapse
|
88
|
Smith RE, Aapro MS, Ludwig H, Pintér T, Šmakal M, Ciuleanu TE, Chen L, Lillie T, Glaspy JA. Darbepoetin Alfa for the Treatment of Anemia in Patients With Active Cancer Not Receiving Chemotherapy or Radiotherapy: Results of a Phase III, Multicenter, Randomized, Double-Blind, Placebo-Controlled Study. J Clin Oncol 2008; 26:1040-50. [PMID: 18227526 DOI: 10.1200/jco.2007.14.2885] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The efficacy and safety of darbepoetin alfa (DA) for treating patients with active cancer and anemia not receiving or planning to receive cytotoxic chemotherapy or myelosuppressive radiotherapy was evaluated. Patients and Methods Patients with active cancer and anemia not receiving or planning to receive chemotherapy or radiotherapy were enrolled onto a phase III, multicenter, randomized, placebo-controlled study and administered placebo or DA 6.75 μg/kg every 4 weeks (Q4W) for up to 16 weeks with a 2-year follow-up for survival. Patients who completed 16 weeks of treatment could receive the same treatment as randomized Q4W for an additional 16 weeks. The primary end point was all occurrences of transfusions from weeks 5 through 17; safety end points included incidence of adverse events and survival. Results The incidence of transfusions between weeks 5 and 17 was lower in the DA group but was not statistically significantly different from that of placebo. DA was associated with an increased incidence of cardiovascular and thromboembolic events and more deaths during the initial 16-week treatment period. Long-term survival data demonstrated statistically significantly poorer survival in patients treated with DA versus placebo (P = .022). This effect varied by baseline covariates including, sex, tumor type, and geographic region; statistical significance diminished (P = .12) when the analysis was adjusted for baseline imbalances or known prognostic factors. Conclusion DA was not associated with a statistically significant reduction in transfusions. Shorter survival was observed in the DA arm; thus, this study does not support the use of erythropoiesis-stimulating agents in this subset of patients with anemia of cancer.
Collapse
Affiliation(s)
- Robert E. Smith
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - Matti S. Aapro
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - Heinz Ludwig
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - Tamás Pintér
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - Martin Šmakal
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - Tudor E. Ciuleanu
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - Li Chen
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - Tom Lillie
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| | - John A. Glaspy
- From the South Carolina Oncology Associates, Columbia, SC; Amgen Inc, Thousand Oaks; University of California, Los Angeles, School of Medicine, Los Angeles, CA; Clinique de Genolier, Genolier, Switzerland; Wilhelminenspital, 1.Medizinische Abteilung mit Onkologie, Vienna, Austria; Petz Aladar County Teaching Hospital, Gyor, Hungary; Ustav Onkologie a Pneumologie Na Plesi, Onkologicke oddeleni, Plesi, Czech Republic; and Institutul Oncologic “Ion Chiricuta,” Cluj-Napoca, Romania
| |
Collapse
|
89
|
|
90
|
|
91
|
Montoya VP, Xie J, Williams D, Woodman RC, Wilhelm FE. An extended maintenance dosing regimen of epoetin alfa 80,000 U every 3 weeks in anemic patients with cancer receiving chemotherapy. Support Care Cancer 2007; 15:1385-92. [PMID: 17541653 DOI: 10.1007/s00520-007-0263-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2006] [Accepted: 04/12/2007] [Indexed: 11/30/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate the safety and efficacy of epoetin alfa (EPO) at an initial dose of 60,000 Units (U) once weekly (QW) followed by extended dosing of 80,000 U every 3 weeks (Q3W) in patients with chemotherapy-induced anemia (CIA). MATERIALS AND METHODS Anemic patients (hemoglobin [Hb] < or = 11 g/dl) receiving Q3W chemotherapy for nonmyeloid malignancy were enrolled in this prospective, open-label, single-arm study to receive EPO 60,000 U subcutaneously (SC) QW (initial dosing phase [IDP]) until a target Hb level of 12 g/dl was reached (maximum 12 weeks). Patients who achieved an Hb level of 12 g/dl at any point during the IDP then entered the extended dosing phase (EDP; EPO 80,000 U SC Q3W). Maximum study duration (IDP + EDP) was 24 weeks. The primary endpoint was the proportion of patients achieving a hematopoietic response (Hb increase > or = 2 g/dl from baseline or Hb > or = 12 g/dl) during the IDP. RESULTS One hundred fifteen patients were enrolled. During the IDP, 76% (84/110) of patients achieved a hematopoietic response, and 15% (17/115) received red blood cell (RBC) transfusion. Sixty-three percent (73/115) of patients entered the EDP, and 88% (64/73) of these patients maintained a mean Hb level > 11.0 and < or =13.0 g/dl. Two of 73 patients received RBC transfusion during the EDP. Adverse events were consistent with the underlying disease and chemotherapy treatment. CONCLUSION These results suggest that initiation of EPO 60,000 U SC QW is effective in the treatment of CIA and that EPO 80,000 U SC Q3W can be an effective extended dosing option.
Collapse
|
92
|
Rohrer Bley C, Wergin M, Roos M, Grenacher B, Kaser-Hotz B. Interrelation of directly measured oxygenation levels, erythropoietin and erythropoietin receptor expression in spontaneous canine tumours. Eur J Cancer 2007; 43:963-7. [PMID: 17254768 DOI: 10.1016/j.ejca.2006.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Revised: 12/06/2006] [Accepted: 12/11/2006] [Indexed: 10/23/2022]
Abstract
The expression of the hypoxia-inducible protein erythropoietin in tumour cells correlates with levels of tumour hypoxia. Our aim was to look for an interrelation of directly measured oxygenation levels, the presence of tissue erythropoietin and its receptor. Data of tumour oxygenation status, plasma and tissue erythropoietin and its receptor in a group of spontaneously occurring tumours in 15 dogs were collected. Polarographic tumour oxygen partial pressure measurements were obtained and data were correlated. Significant positive correlations were found between tissue erythropoietin and the percentages of pO2 values < or = 10 mmHg. Multivariate analysis revealed no parameters influencing plasma erythropoietin levels. Our results show that a co-expression of erythropoietin receptor and its ligand in spontaneous canine tumours exists, that the level of hypoxia in tumour cells correlates with the level of tissue erythropoietin and suggest the need to be quantitatively and functionally tested as novel prognostic biological parameters in neoplastic tissues.
Collapse
Affiliation(s)
- Carla Rohrer Bley
- Section of Diagnostic Imaging and Radiation Oncology, Vetsuisse Faculty, University of Zurich, Winterhurerstrasse 260, CH-8057 Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
93
|
Stockmann C, Fandrey J. Hypoxia-induced erythropoietin production: a paradigm for oxygen-regulated gene expression. Clin Exp Pharmacol Physiol 2006; 33:968-79. [PMID: 17002676 DOI: 10.1111/j.1440-1681.2006.04474.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mechanisms controlling the expression of the gene encoding for the hormone erythropoietin (EPO) are exemplary for oxygen-regulated gene expression. In humans and other mammals, hypoxia modulates EPO levels by increasing expression of the EPO gene. An association between polycythaemia and people living at high altitudes was first reported more than 100 years ago. Since the identification of EPO as the humoral regulator of red blood cell production and the cloning of the EPO gene, considerable progress has been made in understanding the regulation of EPO gene expression. This has finally led to the identification of a widespread cellular oxygen-sensing mechanism. Central to this mechanism is the transcription factor complex hypoxia-inducible factor (HIF)-1. The abundance and activity of HIF-1, a heterodimer of an alpha- and beta-subunit, is predominantly regulated by oxygen-dependent post-translational hydroxylation of the alpha-subunit. Non-heme ferrous iron containing hydroxylases use dioxygen and 2-oxoglutarate to specifically target proline and an asparagine residue in HIF-1alpha. As such, the three prolyl hydroxylases (prolyl hydroxylase domain-containing protein (PHD) 1, PHD2 and PHD3) and the asparagyl hydroxylase (factor inhibiting HIF (FIH)-1) act as cellular oxygen sensors. In addition to erythropoiesis, HIF-1 regulates a broad range of physiologically relevant genes involved in angiogenesis, apoptosis, vasomotor control and energy metabolism. Therefore, the HIF system is implicated in the pathophysiology of many human diseases. In addition to the tight regulation by oxygen tension, temporal and tissue-specific signals limit expression of the EPO gene primarily to the fetal liver and the adult kidney.
Collapse
|
94
|
Lai SY, Grandis JR. Understanding the presence and function of erythropoietin receptors on cancer cells. J Clin Oncol 2006; 24:4675-6. [PMID: 17028292 DOI: 10.1200/jco.2006.08.1190] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
95
|
Constante M, Jiang W, Wang D, Raymond VA, Bilodeau M, Santos MM. Distinct requirements for Hfe in basal and induced hepcidin levels in iron overload and inflammation. Am J Physiol Gastrointest Liver Physiol 2006; 291:G229-37. [PMID: 16565419 PMCID: PMC2891007 DOI: 10.1152/ajpgi.00092.2006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepcidin is a negative regulator of iron absorption produced mainly by the liver in response to changes in iron stores and inflammation, and its levels have been shown to regulate the intestinal basolateral iron transporter ferroportin1 (Fp1). Hereditary hemochromatosis patients and Hfe-deficient mice show inappropriate expression of hepcidin but, in apparent contradiction, still retain the ability to regulate iron absorption in response to alterations of iron metabolism. To further understand the molecular relationships among Hfe, hepcidin, and Fp1, we investigated hepcidin and Fp1 regulation in Hfe-deficient mice (Hfe-/- and beta2m-/-) in response to iron deprivation, iron loading, and acute inflammation. We found that whereas basal hepcidin levels were manifestly dependent on the presence of Hfe and on the mouse background, all Hfe-deficient mice were still able to regulate hepcidin in situations of altered iron homeostasis. In the liver, Fp1 was modulated in opposite directions by iron and LPS, and its regulation in Hfe-deficient mice was similar to that observed in wild-type mice. In addition, we found that iron-deprived mice were able to mount a robust response after LPS challenge and that Toll-like receptor 4 (TLR-4)-deficient mice fail to regulate hepcidin expression in response to LPS. In conclusion, these results suggest that although Hfe is necessary for the establishment of hepcidin basal levels, it is dispensable for hepcidin regulation through both the iron-sensing and inflammatory pathways, and hepatic Fp1 regulation is largely independent of hepcidin and Hfe. The inflammatory pathway overrides the iron-sensing pathway and is TLR-4 dependent.
Collapse
Affiliation(s)
- Marco Constante
- Centre de recherche, Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, Canada
| | | | | | | | | | | |
Collapse
|
96
|
Bohlius J, Weingart O, Trelle S, Engert A. Cancer-related anemia and recombinant human erythropoietin--an updated overview. ACTA ACUST UNITED AC 2006; 3:152-64. [PMID: 16520805 DOI: 10.1038/ncponc0451] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Accepted: 01/15/2006] [Indexed: 01/17/2023]
Abstract
For cancer patients, anemia can be a debilitating problem that negatively influences their overall quality of life and worsens their prognosis. The condition is caused either by the cancer itself or by cytotoxic treatment. Anemia is the primary indication for transfusion of red blood cells, but the development of recombinant human erythropoietins (epoetins) provides an alternative to red blood cell transfusions. Treatment with epoetins has been shown to reduce transfusion rates and increase hemoglobin response. There is some evidence that epoetins improve quality of life. It remains unclear, however, whether erythropoietin affects tumor growth and survival, and this area requires further investigation. Data from clinical trials suggest that erythropoietin increases the risk of thromboembolic complications. In the management of anemic patients, physicians should follow closely the dosing recommendations in products' package inserts or the ASCO/American Society of Hematology guidelines. Treatment of patients beyond the correction of anemia, however, has to be regarded as experimental and is potentially harmful, so should only be conducted in clinical trials.
Collapse
Affiliation(s)
- Julia Bohlius
- Cochrane Haematological Malignancies Group, University of Cologne, Cologne, Germany
| | | | | | | |
Collapse
|
97
|
Laftah A, Sharma N, Brookes M, McKie A, Simpson R, Iqbal T, Tselepis C. Tumour necrosis factor alpha causes hypoferraemia and reduced intestinal iron absorption in mice. Biochem J 2006; 397:61-7. [PMID: 16566752 PMCID: PMC1479761 DOI: 10.1042/bj20060215] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cytokines are implicated in the anaemia of chronic disease by reducing erythropoiesis and increasing iron sequestration in the reticuloendotheial system. However, the effect of cytokines, in particular TNFalpha (tumour necrosis factor alpha), on small bowel iron uptake and iron-transporter expression remains unclear. In the present study, we subjected CD1 male mice to intraperitoneal injection with TNFalpha (10 ng/mouse) and then examined the expression and localization of DMT1 (divalent metal transporter 1), IREG1 (iron-regulated protein 1) and ferritin in duodenum. Liver and spleen samples were used to determine hepcidin mRNA expression. Changes in serum iron and iron loading of duodenum, spleen and liver were also determined. We found a significant (P<0.05) fall in serum iron 3 h post-TNFalpha exposure. This was coincident with increased iron deposition in the spleen. After 24 h of exposure, there was a significant decrease in duodenal iron transfer (P<0.05) coincident with increased enterocyte ferritin expression (P<0.05) and re-localization of IREG1 from the basolateral enterocyte membrane. Hepatic hepcidin mRNA levels remained unchanged, whereas splenic hepcidin mRNA expression was reduced at 24 h. In conclusion, we provide evidence that TNFalpha may contribute to anaemia of chronic disease by iron sequestration in the spleen and by reduced duodenal iron transfer, which seems to be due to increased enterocyte iron binding by ferritin and a loss of IREG1 function. These observations were independent of hepcidin mRNA levels.
Collapse
Affiliation(s)
- Abas H. Laftah
- *Division of Medical Sciences, University of Birmingham, Birmingham B15 2TT, U.K
| | - Naveen Sharma
- *Division of Medical Sciences, University of Birmingham, Birmingham B15 2TT, U.K
| | - Matthew J. Brookes
- *Division of Medical Sciences, University of Birmingham, Birmingham B15 2TT, U.K
| | - Andrew T. McKie
- †Nutrition Research Division, King's College London, London SE1 9NH, U.K
| | - Robert J. Simpson
- †Nutrition Research Division, King's College London, London SE1 9NH, U.K
| | - Tariq H. Iqbal
- ‡Gastroenterology Unit, Walsgrave Hospital, Clifford Bridge Road, Coventry CV2 2DX, U.K
| | - Chris Tselepis
- *Division of Medical Sciences, University of Birmingham, Birmingham B15 2TT, U.K
- To whom correspondence should be addressed (email )
| |
Collapse
|
98
|
Henry DH, Gordan LN, Charu V, Wilhelm FE, Williams D, Xie J, Woodman RC. Randomized, open-label comparison of epoetin alfa extended dosing (80 000 U Q2W) vs weekly dosing (40 000 U QW) in patients with chemotherapy-induced anemia. Curr Med Res Opin 2006; 22:1403-13. [PMID: 16834839 DOI: 10.1185/030079906x115559] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE This randomized, open-label, multicenter study compared the efficacy and safety of epoetin alfa (EPO) 80 000 U every 2 weeks (Q2W) to the FDA-approved regimen of 40 000 U weekly (QW) in patients with chemotherapy-induced anemia. RESEARCH DESIGN AND METHODS A total of 310 patients with nonmyeloid malignancy and baseline hemoglobin (Hb) <or= 11 g/dL who were scheduled to receive chemotherapy for a minimum of 12 weeks were randomized to EPO Q2W or QW for up to 12 weeks, with dose modification to maintain Hb at approximately 12 g/dL. Efficacy analyses used the per-protocol population (patients who completed the study with a value for Hb change) for the primary endpoint only and the modified intent-to-treat (mITT) population (patients who received study drug and had at least one postbaseline Hb value) for the primary and secondary endpoints. RESULTS Analysis of the primary endpoint revealed that the mean change in Hb from baseline to study end was comparable between the Q2W and QW groups in the per-protocol population (1.6 g/dL vs 1.8 g/dL, respectively; treatment difference, -0.2 g/dL; one-sided 95% confidence interval [-0.56, -]); similar results were observed in the mITT population. Among patients on study at Day 29, 9.6% (13/135) and 11.1% (14/126) of patients in the Q2W and QW groups, respectively, received a transfusion between Day 29 and the end of the study (p = 0.709). Dose withholds (21% vs 42%, p < 0.001) and dose reductions (41% vs 59%, p = 0.003) were less common for Q2W than QW. Safety profiles were similar between groups; clinically relevant thrombotic vascular events occurred in 8% of patients in each group. The open-label dosing and the patient attrition rate did not appear to influence overall study results. CONCLUSIONS Extended dosing (80 000 U Q2W) and once-weekly dosing (40 000 U QW) of EPO provided comparable safety and efficacy for chemotherapy-induced anemia.
Collapse
Affiliation(s)
- David H Henry
- Joan Karnell Cancer Center, Pennsylvania Hospital, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
99
|
|
100
|
Wojchowski DM, Menon MP, Sathyanarayana P, Fang J, Karur V, Houde E, Kapelle W, Bogachev O. Erythropoietin-dependent erythropoiesis: New insights and questions. Blood Cells Mol Dis 2006; 36:232-8. [PMID: 16524748 DOI: 10.1016/j.bcmd.2006.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 01/11/2006] [Indexed: 11/19/2022]
Abstract
Committed erythroid progenitor cells require exposure to erythropoietin (Epo) for their survival and for their quantitatively regulated transition to red blood cells. With regard to Epo signal transduction mechanisms, much has been learned from analyses in cell line models, fetal liver or spleen-derived primary erythroblasts and human CD34pos progenitor cells from cord blood or mobilized bone marrow. Presently, we have developed an ex vivo system that efficiently supports the expansion and development of murine adult bone-marrow-derived erythroid progenitor cells. This system is outlined together with its demonstrated utility in studying (for the first time) the signaling capacities of two knocked-in phosphotyrosine-deficient Epo receptor alleles (EpoR-H and EpoR-HM). Ways in which these studies advance an understanding of core Epo signal transduction events are outlined. Also introduced are two new putative negative regulators of Epo-dependent erythropoiesis, DYRK3 and DAPK2 kinases.
Collapse
Affiliation(s)
- Don M Wojchowski
- Maine Medical Center Research Institute and Program in Stem Cell Biology and Regenerative Medicine, ME 04074-7205, USA.
| | | | | | | | | | | | | | | |
Collapse
|