Erythropoietin Biology in Cancer - Clinical Cancer Research

4 downloads 84 Views 501KB Size Report
Jan 15, 2006 - Oncology, Duke University Medical Center, Durham, North Carolina. Received 8/12/05; ...... in advanced squamous cell carcinoma of the head and neck. ... Batra S, Perelman N, Luck LR, Shimada H, Malik P. Pediatric tumor ...
Review

Erythropoietin Biology in Cancer Matthew E. Hardee,1 Murat O. Arcasoy,2 Kimberly L. Blackwell,2,3 John P. Kirkpatrick,3 and Mark W. Dewhirst3

Abstract

Erythropoietin (Epo) has long been known to be the principal hematopoietic growth factor that regulates cellular proliferation and differentiation along the erythroid lineage. Recent studies have shown that Epo is a pleiotropic cytokine that is proangiogenic and exerts broad tissue-protective effects in diverse nonhematopoietic organs. Recombinant Epo (rEpo) has been widely used in the clinic to prevent or treat malignancy-associated anemia. A series of clinical trials have documented the efficacy of rEpo in reducing RBC transfusion requirements and improving quality of life in cancer patients, and a recent meta-analysis suggested a positive effect on survival. However, two randomized trials reported negative outcomes with rEpo, as patients in the rEpo arm fared worse than their placebo-treated counterparts with respect to progression-free survival. The expression of Epo receptor (EpoR) in cancer cells has raised the possibility that exogenous rEpo may exert direct effects on tumor cells associated with the potential for stimulation of proliferation, inhibition of apoptosis, or modulation of sensitivity to chemoradiation therapy.The presence of an autocrine-paracrine Epo-EpoR system in tumors and potential effects of Epo on tumor microenvironment and angiogenesis are consistent with a complex biology for Epo-EpoR signaling in cancer that requires further research.This review describes Epo and EpoR biology, focusing on the pleiotropic effects of Epo on nonhematopoietic tissues as well as the expression and function of EpoR in cancer cells.

Recombinant human erythropoietin (rEpo) has been used frequently in cancer patient care for the prevention or treatment of anemia associated with cancer. The biological effects of Epo are not limited to the hematopoietic system as many recent studies have shown that Epo is a pleiotropic cytokine that is proangiogenic and also exerts broad tissue-protective effects in diverse nonhematopoietic organs. The ability of rEpo to stimulate physiologic and pathologic angiogenesis and the expression of its cellular receptor EpoR in cancer cells and vascular endothelium have suggested that this hormone may exert direct effects on tumor growth and angiogenesis. Although many clinical trials evaluating the beneficial effects of rEpo in cancer care are positive (1 – 4), and a recent meta-analysis suggests a favorable effect on survival (5), two recent randomized trials reported that patients in the rEpo arm fared worse than their placebo-treated counterparts with respect to progression-free survival (6, 7). This controversy provides an impetus to step back and review the biology of Epo and EpoR, focusing on potential functional consequences of Epo-mediated signaling in nonhematopoietic tissues, including cancer cells.

Expression and Function of Epo and its Receptor Epo is a 30.4 kDa glycoprotein produced primarily in the adult kidney under the control of an oxygen-sensing mechanism and regulates the daily production of 2  1011 RBC in the adult bone marrow to maintain the oxygen-carrying capacity of peripheral blood under physiologic conditions (8). Low tissue oxygen tension induces EPO gene expression through both transcriptional activation and mRNA stabilization (9). The hypoxiadependent up-regulation of EPO is a direct result of hypoxia inducible factor-1 activation, a transcription factor that binds a hypoxia responsive element in the 3V flanking region of the EPO gene. Besides hypoxia, there are several factors that modulate Epo production, such as hypoglycemia, increased intracellular calcium, insulin release, estrogen, androgenic steroids, and various cytokines (8). Localized Epo production has been shown in neural tissue (10), in the female genital tract (11), in placenta (12), and in testis (13). Epo production by erythroid progenitors has been implicated in an autocrine-paracrine mechanism of erythropoiesis regulation (14). In the uterus and oviduct, Epo production is inducible by estrogen (11, 15). The biological effects of Epo in hematopoietic cells are mediated through its binding to EpoR, its specific cell surface receptor. EpoR is a member of the type I cytokine receptor family that includes cellular transmembrane receptors for factors such as granulocyte-colony stimulating factor, many of the interleukins, prolactin, and growth hormone. Epo-EpoR signaling is associated with activation of a cytoplasmic, nonreceptor protein tyrosine kinase Jak2 (16) and the downstream signaling molecule Stat5, a cryptic cytoplasmic transcription factor that plays an important role in the regulation of in vivo erythropoiesis (17). Epo-EpoR – induced signaling through

Authors’ Affiliations: Departments of 1Pathology, 2Medicine, and 3Radiation Oncology, Duke University Medical Center, Durham, North Carolina Received 8/12/05; revised 10/14/05; accepted 11/9/05. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Requests for reprints: Mark W. Dewhirst, Department of Radiation Oncology, Duke University Medical Center, Durham, NC 22710. Phone: 919-684-4180; Fax: 919-684-8718; E-mail: dewhirst@ radonc.duke.edu. F 2006 American Association for Cancer Research. doi: 10.1158/1078-0432.CCR-05-1771

Clin Cancer Res 2006;12(2) January 15, 2006

332

www.aacrjournals.org

Erythropoietin Biology in Cancer

angiogenesis, a process that is essential for tumor progression and metastasis, is not established. The exogenous administration of rEpo has been associated with diverse effects in nonhematopoietic tissues. For instance, expression of EpoR in kidney, muscle cells, and the intestine is associated with the ability of rEpo to induce cellular proliferation (22, 28, 38). In other studies, Epo has proved to be a proangiogenic cytokine in a variety of experimental systems. rEpo stimulates the proliferation and migration of endothelial cells (39), and these proliferative effects are associated with increases in endothelin-1 and cytosolic free calcium (40, 41), as well as tyrosine phosphorylation of intracellular proteins, including Stat5 (42). Ribatti et al. (43) reported that the proangiogenic activity of Epo was similar to that of fibroblast growth factor-2 in the chick chorioallontoic membrane assay, and Jaquet et al. (44) found the angiogenic potential of Epo to be similar to that of vascular endothelial growth factor when stimulating human adult myocardial endothelial cells. rEpo has emerged as a major tissue-protective cytokine as many studies have documented that administration of rEpo modulates the physiologic response to various forms of tissue injury (45). In the central nervous system, Epo improves neuronal survival by decreasing neuronal apoptosis and inflammation, exhibiting a protective effect in a variety of injury models, including cerebral ischemia, trauma, and excitotoxins (46). Furthermore, a clinical trial of rEpo in acute ischemic stroke patients showed improvements in clinical recovery scores and outcomes (47). In the cardiovascular system, rEpo protects cardiac myocytes against ischemic injury in a number of different preclinical model systems (48). rEpo has also been shown to protect the kidney from both ischemic and toxic injuries (49, 50) as well as exhibiting significant therapeutic value in a rodent model of diabetic neuropathy (51). Recent reports describing Epo variants, including asialo-Epo (52) and carbamylated Epo, that retain the protective effects of Epo in nonhematopoietic tissues while exhibiting no effect on hematopoietic cells have suggested the presence of fundamental mechanistic differences between Epo-mediated cellular signaling in hematopoietic versus nonhematopoietic cells (53, 54). The tissue-protective effects of Epo in the brain and heart seem to require a second distinct receptor component, the hc-subunit of the interleukin-3 receptor, also shared by interleukin-5 and granulocyte-macrophage colony stimulating factor. In one study, a physical association between hc-subunit and EpoR was reported in coimmunoprecipitation experiments (55). The exact structure of the cell surface receptor complex that mediates the biological effects of Epo in nonhematopoietic cells remains to be characterized. The availability of tissueprotective Epo variants devoid of erythropoietic stimulatory effects may prove clinically useful by abrogating concerns over undesirable elevation of the RBC mass.

pathways other than Jak-Stat, such as phosphatidylinositol 3-kinase-Akt, has been associated with primary erythroblast survival signaling (18), and the activation of the mitogenactivated protein kinase pathway is required for synergistic expansion of erythroid progenitor and precursor cells in response to Epo and stem cell factor (19). Functional EpoR expression has been documented in many nonhematopoietic cell types, including vascular endothelial cells (20), smooth muscle cells (21), skeletal myoblasts (22), cardiac myocytes (23), neurons (24), retinal photoreceptors (25), liver stromal cells (26), placenta (27), kidney (28), and macrophages (29). EpoR expression and signaling in hematopoietic tissues is essential for normal mammalian erythropoiesis during development. Targeted disruption of the genes encoding either EPO or EPOR in mice has revealed that knockout mouse embryos die in utero due to defective fetal liver erythropoiesis (30). EPO or EPOR knockout embryos also exhibit defects in angiogenesis (31) and cardiac morphogenesis with increased apoptosis in endocardium and myocardium (32). However, a recent study in which erythroid EpoR expression was rescued showed that EPOR expression is not required for normal heart development in the mouse (33). Abnormal regulation of Epo-EpoR signaling in hematopoietic cells has been associated with proliferative disorders of the bone marrow characterized by primary erythrocytosis. Primary familial and congenital polycythemia, a disorder characterized by erythrocytosis and Epo hypersensitivity of erythroid progenitor cells, may occur as a consequence of dominant gain-offunction mutations in EpoR. Hematopoietic cells that express these naturally occurring, mutant EpoRs exhibit increased Epo sensitivity associated with deregulation of the rates of Jak2 and Stat5 inactivation (34). More recently, constitutive activation of Jak2 as a consequence of a missense mutation in its pseudokinase domain has been shown to underlie the Epo-independent erythropoiesis that is characteristic of polycythemia vera, a myeloproliferative disorder that results in erythrocytosis (35).

Epo as a Pleiotropic Cytokine Distinct from its essential role in the regulation of RBC production, Epo is involved in diverse nonhematopoietic biological functions. Epo-EpoR signaling is important for physiologic angiogenesis in the developing mouse embryo (31, 32). Furthermore, a functional Epo-EpoR system has been documented in the female genital tract where endogenous EpoEpoR signaling was implicated in regulation of physiologic cyclic uterine angiogenesis (11). In another study, Epo-EpoR signaling was shown to be involved in the physiologic wound healing cascade by promoting angiogenesis and granulation tissue formation, an effect that was inhibited by neutralizing anti-Epo antibodies and recombinant soluble EpoR, an antagonist of Epo-EpoR signaling (29). In the central nervous system, where Epo is produced by astrocytes and EpoR is expressed by neurons, administration of soluble EpoR was associated with augmentation of ischemic brain injury in rats, demonstrating that Epo plays an important role in the response of the brain to neuronal injury as part of an innate response to stressors (36). A recent study showed that Epo-EpoR signaling is a potent angiogenic factor involved in the pathologic angiogenesis of diabetic retinopathy (37). The potential role of Epo in tumor

www.aacrjournals.org

Malignancy-Related Anemia and rEpo Treatment Anemia is an independent prognostic factor for survival in patients with cancer (56). The European Cancer Anaemia Study recently reported that 72% of patients with hematologic malignancies and 66% of solid tumor patients were anemic at some point during the course of their disease although the exact prevalence of anemia varies according to the type of neoplasm

333

Clin Cancer Res 2006;12(2) January 15, 2006

Review

and surgery (60). Although a direct relationship between acute anemia and intratumoral hypoxia has been reported (61), the effect of chronic anemia is more difficult to interpret (62). Brizel et al. (63) found only a weak correlation between anemia and poor tumor oxygenation in head and neck cancer patients and many nonanemic patients had hypoxic tumors. Tumor hypoxia is common among a wide variety of

(57). The pathophysiology of malignancy-related anemia can be multifactorial. In addition to anemia of chronic disease, nutritional deficiencies, bleeding, hemolysis, bone marrow involvement with malignant cells, and chemoradiotherapy can all contribute to anemia. There is evidence that anemia may be associated with a diminished response to radiotherapy (58), chemotherapy (59),

Table 1. Expression of Epo and EpoR in primary tumors and tumor cell lines Primary tumor or cell line name Bladder HBTPL-1, RT112 Breast Primary breast tumors Primary invasive lobular and ductal Primary mammary carcinoma in situ Primary invasive mammary carcinoma MCF7 MDA-MD-231 T47D BT-549 MDA-134 Female reproductive tract Primary cervical SCC Primary cervical ADC Primary endometrial ADC Primary endometrial carcinoma Primary ovarian ADC HeLa (uterine ADC) SKOV3, OVCAR-3 (ovarian) Gastrointestinal tract Primary gastric adenocarcinoma Primary colon tumor 170, 220 (esophageal ADC) AZ521 (gastric carcinoma) SCH (stomach choriocarcinoma) DLD-1,WiDr (colon ADC) HT-29, CX-1 (colon) Head and neck Primary head and neck tumors JHU-022SCC, UM-SCC-22B, UM-22A, UM-22B, PCI-37A, PCI-37B, 1483, PCI-15B Hematologic K562, UT7 (leukemia) HL-60 (promyelocytic leukemia) K562 (erythroleukemia) KG-1a (acute myelogenous leukemia) Kidney Primary renal cell carcinoma tumors Primary Wilms tumor Caki-2, 786-0 (renal cell carcinoma) KTCTL-26, KTCTL-103, KTCTL-30 (kidney carcinoma)

Method; EpoR/Epo status

Reference

RT-PCR,WB; EpoR+

(83)

IHC; EpoR+, Epo+ IHC; EpoR+, Epo+ IHC; EpoR+, Epo+ IHC; EpoR+, Epo+ RT-PCR,WB; EpoR+ RT-PCR,WB; EpoR+ RT-PCR,WB; EpoR+, Epo+ WB; EpoR+ WB; EpoR+

(88, 79) (77) (96) (96) (79, 88) (77, 88) (77, 87) (77) (77)

RT-PCR; EpoR+, Epo+ RT-PCR; EpoR+ RT-PCR; EpoR+, Epo+ IHC; EpoR+, Epo+ RT-PCR; EpoR+, Epo+ RT-PCR; EpoR+, Epo+ WB, RT-PCR; EpoR+

(86, 97) (97) (86, 97) (98) (86, 97) (83, 87) (79)

IHC; EpoR+ RT-PCR,WB; EpoR+ RT-PCR; EpoR+, Epo+ RT-PCR; EpoR+, Epo+ RT-PCR; EpoR+, Epo+ RT-PCR; EpoR+, Epo+ RT-PCR, IHC,WB; EpoR+

(83, 99) (79) (87) (87) (87) (79, 87) (78)

IHC; EpoR+, Epo+ RT-PCR,WB; EpoR+, Epo+

(82, 100) (81, 82)

RT-PCR; EpoR+, Epo+ RT-PCR,WB; EpoR+ RT-PCR,WB; EpoR+ RT-PCR,WB; EpoR+

(87) (83) (83) (83)

RT-PCR; EpoR+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR; EpoR+ RT-PCR,WB; EpoR+

(80) (78) (80) (83)

(Continued on the following page)

Clin Cancer Res 2006;12(2) January 15, 2006

334

www.aacrjournals.org

Erythropoietin Biology in Cancer

anemic rats bearing mammary adenocarcinoma flank tumors improved tumor oxygenation independent of the effects on hemoglobin levels. Other studies have similarly shown a decrease in the hypoxic fraction of solid tumors with systemic rEpo treatment: one in an i.p. hemorrhagic ascites model of anemia in mice (73) and another in a total body irradiation anemia model in rats in which radiotherapy efficacy was also increased by rEpo administration (67). Epo alone was not

malignancies and is a factor associated with treatment resistance, aggressive clinical phenotype, and poor prognosis (64, 65). Beyond correcting anemia in preclinical anemic tumor models, rEpo has been shown to restore radiosensitivity, increase tumor growth delay (66 – 70), and increase cytotoxicity of chemotherapeutics (71). In a preclinical model, Blackwell et al. (72) found that systemic rEpo administration to non-

Table 1. Expression of Epo and EpoR in primary tumors and tumor cell lines (Cont’d) Primary tumor or cell line name

Method; EpoR/Epo status

Liver Primary fetal hepatoblastoma Primary embryonal hepatoblastoma HepG2, PLC (hepatoma) Lung Primary lung tumor Primary lung adenocarinoma Primary lung SCC A549 (lung ADC) SBC3 (small cell carcinoma) Melanoma Wm35, wm3211, sbcl2 (radial growth phase) Wm793, wm1366, wm298 (vertical growth phase) Wm9, 1205Lu, 451lu (metastatic melanoma) G361, P39, P22, C32TG Nervous system Primary pilocytic astrocytoma Primary anaplastic ependymoma Primary medulloblastoma Primary neuroblastoma T98G, A172 (glioblastoma) CHLA-90, SK-N-RA, KCNR, LHN, CHLA-15, CHLA-20, SK-N-FI, SK-N-BE-2, CHLA-171, SAN, LAN-5, LAN-6, CHLA-134 (neuroblastoma) DAOY (medulloblastoma) Pancreatic MIA PaCa Dan-G Prostate Primary adenocarcinoma PC-3 LNCaP DU145 Sarcoma Primary ESFT Primary embryonal rhabdomyosarcoma Primary alveolar rhabdomyosarcoma CHP-100, A5838, SK-N-MC, TC-106,TC-32,TC-71 (ESFT) RD (rhabdomyosarcoma) Thyroid Primary papillary thyroid carcinomas S117 (thyroid sarcoma)

Reference

RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR,WB; EpoR+, Epo+

(78) (78) (83, 87)

RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR; EpoR+, Epo+ RT-PCR,WB; EpoR+

(79) (101) (101) (87) (87)

WB; EpoR+, Epo+ WB; EpoR+, Epo+ WB; EpoR+, Epo+ RT-PCR; EpoR+, Epo+

(89) (89) (89) (87)

RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR; EpoR+, Epo+ RT-PCR, IHC; EpoR+

(78) (78) (78) (78) (87) (78)

RT-PCR, IHC; EpoR+

(78)

RT-PCR; EpoR+, Epo+ RT-PCR,WB; EpoR+

(87) (83)

IHC; EpoR+, Epo+ RT-PCR,WB; EpoR+, Epo+ RT-PCR,WB; EpoR+, Epo+ RT-PCR,WB; EpoR+

(102) (87, 102) (102) (83)

RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+, Epo+ RT-PCR, IHC; EpoR+

(78) (78) (78) (78)

RT-PCR; EpoR+

(83)

IHC; EpoR+, Epo+/RT-PCR,WB; EpoR+

(103) (83)

Abbreviations: SCC, squamous cell carcinoma; ADC, adenocarcinoma; ESFT, Ewing sarcoma family tumor. RT-PCR, reverse transcription-PCR; IHC, immunohistochemistry; WB,Western blot.

www.aacrjournals.org

335

Clin Cancer Res 2006;12(2) January 15, 2006

Review

monolayer cultures of MCF7 and BT-549 breast cancer cells. Similar findings were reported with Caki-2, 786-0, and RAG renal cell carcinoma cells (80). A limited rEpo-mediated induction of proliferation and stimulation of invasion was reported in human head and neck squamous cell carcinoma cell lines (81). Further, Mohyeldin et al. (82) reported a correlation between disease progression and EpoR expression in head and neck cancer biopsies, and an increase in invasiveness in head and neck cancer cells in response to rEpo. In contrast, however, Westphal et al. (83) reported no proliferative effects of exogenous rEpo on six EpoR-positive tumor cell lines, and Selzer et al. (84) reported that EpoRpositive human melanoma cells did not proliferate in response to rEpo. Similarly, in a study of 22 tumor cell lines derived from a range of human solid tumors, rEpo exerted no growthmodulating effects (85). Taken together, these findings suggest that exogenous Epo does not consistently provide a proliferative stimulus for cancer cells. Whether the contrasting data with respect to the proliferative effects of Epo are due to differences in the cancer cell types tested or the experimental models is unclear. Further studies in xenograft models of human cancer will be required to evaluate the effects of Epo on tumor growth rates in the presence or absence of concomitant chemoradiotherapy. There is an accumulating body of experimental evidence for the presence of functional endogenous Epo-EpoR signaling in tumors from studies that used strategies to block Epo signaling pathways. Yasuda et al. (86) reported that Epo signaling contributes to the growth and angiogenesis of female reproductive tract tumors. Blockade of Epo signaling with local soluble EpoR or anti-Epo antibody resulted in tumor cell destruction and reduction of vascularity in ovarian and uterine cancer xenografts, associated with an increase in apoptotic death of both tumor cells and vascular endothelial cells. It was also shown that i.p. injections of an EpoR antagonist blocked Stat5 phosphorylation and inhibited melanoma and stomach choriocarcinoma tumor cell survival and angiogenesis (87). In another study, Arcasoy et al. (88) found that administration of Epo-EpoR inhibitors, including neutralizing anti-Epo antibody, soluble EpoR, or pharmacologic Jak2 inhibitor in rat mammary adenocarcinoma tumors in a tumor chamber model, resulted in significant tumor growth delay. These preclinical data suggest that the exploration of strategies to block Epo-EpoR function to target tumor growth and angiogenesis may be warranted. Several studies reported Epo-mediated modulation of tumor cell sensitivity to apoptosis and investigated the ability of Epo to influence the effects of chemoradiotherapy with contrasting results, in both in vitro or in vivo experimental models. Batra et al. (78) showed increased expression of antiapoptotic genes (bcl-xL, bcl-2, and mcl-1), as well as an increase in nuclear factor-nB DNA binding activity in Ewing sarcoma family tumor and neuroblastoma cell lines in response to rEpo. This finding was associated with increased secretion of proangiogenic cytokines, which promoted endothelial cell proliferation and chemotaxis. In human melanoma cells, incubation with rEpo increased tumor cell resistance to hypoxia-induced cell death (89). These cells also showed increased mitochondrial membrane potential, a possible mechanism for the observed antiapoptotic effect. In a study of HeLa uterine cervix carcinoma cells, Acs et al. (90) reported rEpo-mediated, dose-dependent reduction in apoptosis and increase in the

associated with modulation of microvessel density or tumor growth rate in these studies. The available rEpo preparations include epoetin-a, epoetinh, and the longer-acting darbepoetin-a, and are effective in increasing hemoglobin levels, decreasing the need for red cell transfusions, and improving quality of life in cancer patients receiving therapy (1 – 3, 74). In several clinical trials, improvements in survival were suggested with rEpo administration to anemic cancer patients. In a retrospective study, significant improvements in response, control, and survival rates were observed in patients with squamous cell carcinomas of the oral cavity and oropharynx treated with chemoradiation and rEpo (75). Trends toward improved survival with rEpo treatment of anemic cancer patients were also reported in a randomized, double-blind, placebo-controlled trial of nonmyeloid malignancies (4) and a study of lung cancer patients receiving chemotherapy (74), although the latter was not sufficiently powered to evaluate survival. In contrast, two recent clinical trials reported adverse outcome associated with rEpo therapy of metastatic breast cancer patients undergoing chemotherapy (7) and head and neck cancer patients undergoing radiotherapy (6). The unfavorable survival rate in rEpo-treated patients was associated, at least in part, with disease progression in both trials. Additionally, rEpo administration seemed to be associated with an increased incidence of deep venous thrombosis in the breast cancer trial, as well as in a trial of concurrent chemoradiation in cervical cancer patients (76). Several design- and treatment-related issues have been pointed out in the breast and head-neck cancer trials, making their interpretation difficult. In contrast to the results of these trials, a recent meta-analysis provided suggestive, but inconclusive, evidence that rEpo may improve overall survival in anemic cancer patients (5). Further prospective, randomized, and controlled studies will be required to investigate the effects, if any, of rEpo therapy on disease progression and survival of cancer patients.

Epo and EpoR Expression and Function in Malignant Tumors Several recent studies have reported expression of EpoR in tumor cell lines as well as primary cancers (Table 1). Many tumors were also reported to express Epo mRNA transcripts and protein, suggesting the potential for generation of an autocrine or paracrine growth-stimulatory Epo-EpoR loop in cancer cells. The expression of Epo in tumor cell lines was reported to be hypoxia inducible (77, 78). The expression of EpoR splice variants was also shown in cancer cell lines, which could be important in modulating the cellular effects of Epo (79). Furthermore, the expression of EpoR in vascular endothelium in tumors has suggested potential effects of Epo on the tumor microenvironment, such as the stimulation of tumor angiogenesis. In addition, it has been suggested that the potential for direct activation of Epo-EpoR signaling pathways in cancer cells may be associated with modulation of various aspects of tumor biology, including cellular proliferation, apoptosis, and sensitivity to chemoradiation therapy (Fig. 1). There have been in vitro studies that reported increased proliferation of tumor cells in response to exogenous rEpo. Acs et al. (77) reported a f25% increase in the proliferation of

Clin Cancer Res 2006;12(2) January 15, 2006

336

www.aacrjournals.org

Erythropoietin Biology in Cancer

Fig. 1. Potential effects and mechanisms of Epo-EpoR signaling in tumors. Cancer cells and tumor endothelium express EpoR, illustrated here as a homodimer, analogous to the functional EpoR in erythroid cells.The exact structure of the cell surface receptor complex that mediates the effects of Epo in nonhematopoietic tissues and cancer cells has not been defined. In this hypothetical model, EpoR activation may be stimulated by Epo from the systemic circulation (renal or exogenous source). In addition, coexpression of Epo and EpoR in tumor cells and the expression of EpoR in vascular endothelial cells may result in the generation of an autocrine-paracrine Epo-EpoR activation loop, constituting a potential therapeutic target in tumors where Epo-EpoR signaling may be involved in tumor angiogenesis and progression. The direct, in vivo effects of Epo-EpoR signaling on cellular proliferation, tumor oxygenation, apoptosis, tumor angiogenesis, metastasis, and sensitivity to chemoradiation therapy remain to be characterized.

stimulatory effect on the in vitro proliferation of cancer cells (95). Further well-controlled studies in xenograft models of different types of cancer are required to characterize the ability of rEpo to modulate tumor growth, angiogenesis, chemoradiation sensitivity, and apoptosis.

surviving fraction of tumor cells treated with cisplatin. In another study, overexpression of EpoR in HeLa cells was associated with rEpo-mediated activation of nuclear factor-nB and a 50% increase in plating efficiency of HeLa cells, although this effect was not associated with a change in radiation sensitivity (91). In human melanoma cells, rEpo increased cell viability during treatment with varying concentrations of dacarbazine and cisplatin, although rEpo-induced resistance to dacarbazine was higher than to cisplatin (89). Using U87 human malignant glioma and HT100 primary cervical cancer cell lines, Belenkov et al. (92) reported an increase in both radiation and cisplatin resistance in response to exogenous rEpo, an effect that was reversible upon addition of a Jak2 inhibitor. Although these data suggest that rEpo may protect cancer cells against the effects of chemoradiotherapy, other studies have reported contrasting findings. For instance, human renal cell carcinoma and myelomonocytic leukemia cell lines treated with rEpo exhibited an increase in apoptosis in response to daunorubicin and vinblastine, which was associated with inactivation of the nuclear factor-nB pathway (93). In a preclinical myeloma model, rEpo was associated with stimulation of antitumor immune responses resulting in tumor regression (94). In a more recent study, rEpo administration was associated with remodeling of tumor microvessels and increased chemosensitivity to 5-fluorouracil treatment of human squamous cell and colorectal carcinoma xenografts but without a

www.aacrjournals.org

Summary Epo is a pleiotropic cytokine that exerts diverse biological effects in many nonhematopoietic tissues. Epo is involved in the wound-healing cascade, functions as a proangiogenic cytokine during physiologic angiogenesis in the embryo and uterus, and exerts tissue-protective effects as part of an innate response to stressors. The recent characterization of Epo variants, such as asialo-Epo and carbamylated-Epo, that retain nonhematopoietic, tissue-protective properties of Epo without stimulating erythropoiesis has uncovered new areas of research into mechanisms of Epo-mediated signaling in nonhematopoietic tissues as well as novel clinical applications for rEpo and its derivatives in disorders other than anemia. rEpo has been widely used in the prevention and treatment of cancer-related anemia, leading to increased hemoglobin levels, reduction of RBC transfusion requirements, and improvement of quality of life. Despite these beneficial effects and some studies suggesting improved survival trend in Epo-treated patients with cancer, two recent prospective, randomized clinical trials involving head-neck

337

Clin Cancer Res 2006;12(2) January 15, 2006

Review

to tumor behavior and response to treatment. Further, the expression of EpoR alone is not always sufficient to modulate these pathways as shown by the variable effect of rEpo on chemoradiation sensitivity and the absence of a consistent proliferative effect in different types of cancer cells. The potential development of erythropoiesis-stimulating agents that are devoid of the biological effects of Epo in nonhematopoietic cells may constitute a future strategy for selective targeting of erythropoietic agent therapy. Characterization of the mechanisms of the biological effects of Epo on nonhematopoietic cells, in particular cancer cells, should continue to be explored to optimize the use of Epo in anticancer therapy.

and breast cancer patients have raised concerns over potential adverse effects of rEpo in cancer patients. The expression of EpoR in cancer cells has suggested the possibility that exogenous rEpo may exert direct effects on tumor cells associated with stimulation of proliferation, inhibition of apoptosis, or modulation of sensitivity to chemoradiation therapy. The presence of an autocrine-paracrine Epo-EpoR system in tumors and possible effects of Epo on tumor microenvironment and angiogenesis are consistent with a complex biology for Epo-EpoR signaling in cancer that requires further research. The overall direct effect of Epo-EpoR signaling is clearly not a straightforward one, as signaling can potentially activate several pathways important

References 1. Glaspy J, Bukowski R, Steinberg D, et al. Impact of therapy with epoetin a on clinical outcomes in patients with nonmyeloid malignancies during cancer chemotherapy in community oncology practice. Procrit Study Group. J Clin Oncol 1997;15:1218 ^ 34. 2. Demetri GD, Kris M,Wade J, Degos L, Cella D. Quality-of-life benefit in chemotherapy patients treated with epoetin a is independent of disease response or tumor type: results from a prospective community oncology study. Procrit Study Group. J Clin Oncol 1998;16:3412 ^ 25. 3. Gabrilove JL, Cleeland CS, Livingston RB, et al. Clinical evaluation of once-weekly dosing of epoetin a in chemotherapy patients: improvements in hemoglobin and quality of life are similar to three-times-weekly dosing. J Clin Oncol 2001;19:2875 ^ 82. 4. Littlewood TJ, Bajetta E, Nortier JW, Vercammen E, Rapoport B. Effects of epoetin a on hematologic parameters and quality of life in cancer patients receiving nonplatinum chemotherapy: results of a randomized, double-blind, placebo-controlled trial. J Clin Oncol 2001;19:2865 ^ 74. 5. Bohlius J, Langensiepen S, Schwarzer G, et al. Recombinant human erythropoietin and overall survival in cancer patients: results of a comprehensive meta-analysis. J Natl Cancer Inst 2005;97: 489 ^ 98. 6. Henke M, Laszig R, Rube C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, doubleblind, placebo-controlled trial. Lancet 2003;362: 1255 ^ 60. 7. Leyland-Jones B, Semiglazov V, Pawlicki M, et al. Maintaining normal hemoglobin levels with epoetin a in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. J Clin Oncol 2005; 23:5960 ^ 72. 8. Jelkmann W. Erythropoietin: structure, control of production, and function. Physiol Rev 1992;72: 449 ^ 89. 9. Ebert BL, Bunn HF. Regulation of the erythropoietin gene. Blood 1999;94:1864 ^ 77. 10. Masuda S, Okano M,Yamagishi K, et al. A novel site of erythropoietin production. Oxygen-dependent production in cultured rat astrocytes. J Biol Chem 1994;269:19488 ^ 93. 11. Yasuda Y, Masuda S, Chikuma M, et al. Estrogendependent production of erythropoietin in uterus and its implication in uterine angiogenesis. J Biol Chem 1998;273:25381 ^ 7. 12. Conrad KP, Benyo DF, Westerhausen-Larsen A, Miles TM. Expression of erythropoietin by the human placenta. FASEB J 1996;10:760 ^ 8. 13. Magnanti M, Gandini O, Giuliani L, et al. Erythropoietin expression in primary rat Sertoli and peritubular myoid cells. Blood 2001;98:2872 ^ 4. 14. SatoT, MaekawaT, Watanabe S,Tsuji K, NakahataT.

Erythroid progenitors differentiate and mature in response to endogenous erythropoietin. J Clin Invest 2000;106:263 ^ 70. 15. Masuda S, Kobayashi T, Chikuma M, Nagao M, Sasaki R. The oviduct produces erythropoietin in an estrogen- and oxygen-dependent manner. Am J Physiol Endocrinol Metab 2000;278:E1038 ^ 44. 16. Parganas E, Wang D, Stravopodis D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 1998;93:385 ^ 95. 17. Socolovsky M, Fallon AE, Wang S, Brugnara C, Lodish HF. Fetal anemia and apoptosis of red cell progenitors in Stat5a / 5b / mice: a direct role for Stat5 in Bcl-X(L) induction. Cell 1999;98:181 ^ 91. 18. Myklebust JH, Blomhoff HK, Rusten LS, Stokke T, Smeland EB. Activation of phosphatidylinositol 3-kinase is important for erythropoietin-induced er y thropoiesis from CD3 4(+) hematopoietic progenitor cells. Exp Hematol 2002;30:990 ^ 1000. 19. Arcasoy MO, Jiang X. Co-operative signalling mechanisms required for erythroid precursor expansion in response to erythropoietin and stem cell factor. Br J Haematol 2005;130:121 ^ 9. 20. Anagnostou A, Liu Z, Steiner M, et al. Erythropoietin receptor mRNA expression in human endothelial cells. Proc Natl Acad Sci U S A 1994;91: 3974 ^ 8. 21. Ammarguellat F, Gogusev J, Drueke TB. Direct effect of erythropoietin on rat vascular smooth-muscle cell via a putative erythropoietin receptor. Nephrol Dial Transplant 1996;11:687 ^ 92. 22. Ogilvie M, Yu X, Nicolas-Metral V, et al. Erythropoietin stimulates proliferation and interferes with differentiation of myoblasts. J Biol Chem 2000; 275:39754 ^ 61. 23. Parsa CJ, Kim J, Riel RU, et al. Cardioprotective effects of erythropoietin in the reperfused ischemic heart: a potential role for cardiac fibroblasts. J Biol Chem 2004;279:20655 ^ 62. 24. Nagai A, Nakagawa E, Choi HB, et al. Erythropoietin and erythropoietin receptors in human CNS neurons, astrocytes, microglia, and oligodendrocytes grown in culture. J Neuropathol Exp Neurol 2001;60:386 ^ 92. 25. Grimm C,Wenzel A, Groszer M, et al. HIF-1-induced erythropoietin in the hypoxic retina protects against light-induced retinal degeneration. Nat Med 2002;8: 718 ^ 24. 26. Ohneda O, Yanai N, Obinata M. Erythropoietin as a mitogen for fetal liver stromal cells which support erythropoiesis. Exp Cell Res 1993;208:327 ^ 31. 27. Sawyer ST, Krantz SB, Sawada K. Receptors for erythropoietin in mouse and human erythroid cells and placenta. Blood 1989;74:103 ^ 9. 28.Westenfelder C, Biddle DL, Baranowski RL. Human, rat, and mouse kidney cells express functional erythropoietin receptors. Kidney Int 1999;55:808 ^ 20. 29. Haroon ZA, Amin K, Jiang X, Arcasoy MO. A novel

Clin Cancer Res 2006;12(2) January 15, 2006

338

role for erythropoietin during fibrin-induced woundhealing response. Am J Pathol 2003;163:993 ^ 1000. 30. Wu H, Liu X, Jaenisch R, Lodish HF. Generation of committed erythroid BFU-E and CFU-E progenitors does not require erythropoietin or the erythropoietin receptor. Cell 1995;83:59 ^ 67. 31. Kertesz N,Wu J, ChenTH, Sucov HM,Wu H.The role of erythropoietin in regulating angiogenesis. Dev Biol 2004;276:101 ^ 10. 32. Wu H, Lee SH, Gao J, Liu X, Iruela-Arispe ML. Inactivation of erythropoietin leads to defects in cardiac morphogenesis. Development 1999;126: 3597 ^ 605. 33. Suzuki N, Ohneda O, Takahashi S, et al. Erythroidspecific expression of the erythropoietin receptor rescued its null mutant mice from lethality. Blood 2002; 100:2279 ^ 88. 34. Arcasoy MO, Karayal AF. Erythropoietin hypersensitivity in primary familial and congenital polycythemia: role of tyrosines Y285 and Y344 in erythropoietin receptor cytoplasmic domain. Biochim Biophys Acta 2005;1740:17 ^ 28. 35. James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005;434: 1144 ^ 8. 36. Sakanaka M, WenTC, Matsuda S, et al. In vivo evidence that erythropoietin protects neurons from ischemic damage. Proc Natl Acad Sci U S A 1998; 95:4635 ^ 40. 37. Watanabe D, Suzuma K, Matsui S, et al. Erythropoietin as a retinal angiogenic factor in proliferative diabetic retinopathy. N Engl J Med 2005;353: 782 ^ 92. 38. Juul SE, Ledbetter DJ, Joyce AE, et al. Erythropoietin acts as a trophic factor in neonatal rat intestine. Gut 2001;49:182 ^ 9. 39. Anagnostou A, Lee ES, Kessimian N, Levinson R, Steiner M. Erythropoietin has a mitogenic and positive chemotactic effect on endothelial cells. Proc Natl Acad Sci U S A 1990;87:5978 ^ 82. 40. Carlini RG, Dusso AS, Obialo CI, Alvarez UM, Rothstein M. Recombinant human erythropoietin (rHuEPO) increases endothelin-1 release by endothelial cells. Kidney Int 1993;43:1010 ^ 4. 41. Carlini RG, Reyes AA, Rothstein M. Recombinant human erythropoietin stimulates angiogenesis in vitro. Kidney Int 1995;47:740 ^ 5. 42. Haller H, Christel C, Dannenberg L, et al. Signal transduction of erythropoietin in endothelial cells. Kidney Int 1996;50:481 ^ 8. 43. Ribatti D, Presta M, Vacca A, et al. Human erythropoietin induces a pro-angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. Blood 1999;93:2627 ^ 36. 44. Jaquet K, Krause K, Tawakol-Khodai M, Geidel S, Kuck KH. Erythropoietin and VEGF exhibit equal

www.aacrjournals.org

Erythropoietin Biology in Cancer angiogenic potential. Microvasc Res 2002;64: 326 ^ 33. 45. Maiese K, Li F, Chong ZZ. New avenues of exploration for erythropoietin. JAMA 2005;293:90 ^ 5. 46. Grasso G, Sfacteria A, Cerami A, Brines M. Erythropoietin as a tissue-protective cytokine in brain injury: what do we know and where do we go? Neuroscientist 2004;10:93 ^ 8. 47. Ehrenreich H, Hasselblatt M, Dembowski C, et al. Erythropoietin therapy for acute stroke is both safe and beneficial. Mol Med 2002;8:495 ^ 505. 48. Hanlon PR, Fu P, Wright GL, et al. Mechanisms of erythropoietin-mediated cardioprotection during ischemia-reperfusion injury: role of protein kinase C and phosphatidylinositol 3-kinase signaling. FASEB J 2005;19:1323 ^ 5. 49. Vesey DA, Cheung C, Pat B, et al. Erythropoietin protects against ischaemic acute renal injury. Nephrol Dial Transplant 2004;19:348 ^ 55. 50. Bagnis C, Beaufils H, Jacquiaud C, et al. Erythropoietin enhances recovery after cisplatin-induced acute renal failure in the rat. Nephrol Dial Transplant 2001;16: 932 ^ 8. 51. Bianchi R, Buyukakilli B, Brines M, et al. Erythropoietin both protects from and reverses experimental diabetic neuropathy. Proc Natl Acad Sci U S A 2004;101: 823 ^ 8. 52. Erbayraktar S, Grasso G, Sfacteria A, et al. Asialoerythropoietin is a nonerythropoietic cytokine with broad neuroprotective activity in vivo. Proc Natl Acad Sci U S A 2003;100:6741 ^ 6. 53. Leist M, Ghezzi P, Grasso G, et al. Derivatives of erythropoietin that are tissue protective but not erythropoietic. Science 2004;305:239 ^ 42. 54. Fiordaliso F, Chimenti S, Staszewsky L, et al. A nonerythropoietic derivative of erythropoietin protects the myocardium from ischemia-reperfusion injury. Proc Natl Acad Sci U S A 2005;102: 2046 ^ 51. 55. Brines M, Grasso G, Fiordaliso F, et al. Erythropoietin mediates tissue protection through an erythropoietin and common h-subunit heteroreceptor. Proc Natl Acad Sci U S A 2004;101:14907 ^ 12. 56. Caro JJ, Salas M, Ward A, Goss G. Anemia as an independent prognostic factor for survival in patients with cancer: a systemic, quantitative review. Cancer 2001;91:2214 ^ 21. 57. Ludwig H, Van Belle S, Barrett-Lee P, et al. The European Cancer Anaemia Survey (ECAS): a large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur J Cancer 2004;40: 2293 ^ 306. 58. Nordsmark M, Overgaard M, Overgaard J. Pretreatment oxygenation predicts radiation response in advanced squamous cell carcinoma of the head and neck. Radiother Oncol 1996;41:31 ^ 9. 59. Teicher BA, Holden SA, al-Achi A, Herman TS. Classification of antineoplastic treatments by their differential toxicity toward putative oxygenated and hypoxic tumor subpopulations in vivo in the FSaIIC murine fibrosarcoma. Cancer Res 1990;50: 3339 ^ 44. 60. Hockel M, Schlenger K, Hockel S, et al. Tumor hypoxia in pelvic recurrences of cervical cancer. Int J Cancer 1998;79:365 ^ 9. 61. Hirst DG. What is the importance of anaemia in radiotherapy? The value of animal studies. Radiother Oncol 1991;20 Suppl 1:29 ^ 33. 62. Harrison LB, Chadha M, Hill RJ, Hu K, Shasha D. Impact of tumor hypoxia and anemia on radiation therapy outcomes. Oncologist 2002; 7:492 ^ 508. 63. Brizel DM, Dodge RK, Clough RW, Dewhirst MW. Oxygenation of head and neck cancer: changes during radiotherapy and impact on treatment outcome. Radiother Oncol 1999;53:113 ^ 7. 64. Graeber TG, Osmanian C, Jacks T, et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature 1996;379: 88 ^ 91.

www.aacrjournals.org

65. Vaupel P, Harrison L. Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 2004;9 Suppl 5:4 ^ 9. 66. Thews O, Koenig R, Kelleher DK, Kutzner J, Vaupel P. Enhanced radiosensitivity in experimental tumours following erythropoietin treatment of chemotherapy-induced anaemia. Br J Cancer 1998;78: 752 ^ 6. 67. Ning S, Hartley C, Molineux G, Knox SJ. Darbepoietin a potentiates the efficacy of radiation therapy in mice with corrected or uncorrected anemia. Cancer Res 2005;65:284 ^ 90. 68. Stuben G, Pottgen C, Knuhmann K, et al. Erythropoietin restores the anemia-induced reduction in radiosensitivity of experimental human tumors in nude mice. Int J Radiat Oncol Biol Phys 2003;55: 1358 ^ 62. 69. Pinel S, Barberi-Heyob M, Cohen-Jonathan E, et al. Erythropoietin-induced reduction of hypoxia before and during fractionated irradiation contributes to improvement of radioresponse in human glioma xenografts. Int J Radiat Oncol Biol Phys 2004;59: 250 ^ 9. 70. Stuben G, Thews O, Pottgen C, et al. Recombinant human erythropoietin increases the radiosensitivity of xenografted human tumours in anaemic nude mice. J Cancer Res Clin Oncol 2001;127: 346 ^ 50. 71. Thews O, Kelleher DK, Vaupel P. Erythropoietin restores the anemia-induced reduction in cyclophosphamide cytotoxicity in rat tumors. Cancer Res 2001; 61:1358 ^ 61. 72. Blackwell KL, Kirkpatrick JP, Snyder SA, et al. Human recombinant erythropoietin significantly improves tumor oxygenation independent of its effects on hemoglobin. Cancer Res 2003;63: 6162 ^ 5. 73. Kelleher DK, Mattheinsen U, Thews O, Vaupel P. Blood flow, oxygenation, and bioenergetic status of tumors after erythropoietin treatment in normal and anemic rats. Cancer Res 1996;56:4728 ^ 34. 74. Vansteenkiste J, Pirker R, Massuti B, et al. Double-blind, placebo-controlled, randomized phase III trial of darbepoetin a in lung cancer patients receiving chemotherapy. J Natl Cancer Inst 2002;94: 1211 ^ 20. 75. Glaser CM, Millesi W, Kornek GV, et al. Impact of hemoglobin level and use of recombinant erythropoietin on efficacy of preoperative chemoradiation therapy for squamous cell carcinoma of the oral cavity and oropharynx. Int J Radiat Oncol Biol Phys 2001;50:705 ^ 15. 76.WunT, Law L, Harvey D, et al. Increased incidence of symptomatic venous thrombosis in patients with cervical carcinoma treated with concurrent chemotherapy, radiation, and erythropoietin. Cancer 2003;98: 1514 ^ 20. 77. Acs G, Acs P, Beckwith SM, et al. Erythropoietin and erythropoietin receptor expression in human cancer. Cancer Res 2001;61:3561 ^ 5. 78. Batra S, Perelman N, Luck LR, Shimada H, Malik P. Pediatric tumor cells express erythropoietin and a functional erythropoietin receptor that promotes angiogenesis and tumor cell survival. Lab Invest 2003;83:1477 ^ 87. 79. Arcasoy MO, Jiang X, Haroon ZA. Expression of erythropoietin receptor splice variants in human cancer. Biochem Biophys Res Commun 2003;307: 999 ^ 1007. 80. Westenfelder C, Baranowski RL. Erythropoietin stimulates proliferation of human renal carcinoma cells. Kidney Int 2000;58:647 ^ 57. 81. Lai SY, Childs EE, Xi S, et al. Erythropoietinmediated activation ofJAK-STATsignaling contributes to cellular invasion in head and neck squamous cell carcinoma. Oncogene 2005;24:4442 ^ 9. 82. Mohyeldin A, Lu H, Dalgard C, et al. Erythropoietin signaling promotes invasiveness of human head and neck squamous cell carcinoma. Neoplasia 2005;7: 537 ^ 43. 83. Westphal G, Niederberger E, Blum C, et al. Erythro-

339

poietin and G-CSF receptors in human tumor cells: expression and aspects regarding functionality. Tumori 2002;88:150 ^ 9. 84. Selzer E,Wacheck V, Kodym R, et al. Erythropoietin receptor expression in human melanoma cells. Melanoma Res 2000;10:421 ^ 6. 85. Berdel WE, Oberberg D, Reufi B,Thiel E. Studies on the role of recombinant human erythropoietin in the growth regulation of human nonhematopoietic tumor cells in vitro. Ann Hematol 1991;63:5 ^ 8. 86. YasudaY, MushaT,Tanaka H, et al. Inhibition of erythropoietin signalling destroys xenografts of ovarian and uterine cancers in nude mice. Br J Cancer 2001;84: 836 ^ 43. 87. Yasuda Y, Fujita Y, Matsuo T, et al. Erythropoietin regulates tumour growth of human malignancies. Carcinogenesis 2003;24:1021 ^ 9. 88. Arcasoy MO, Amin K, Karayal AF, et al. Functional significance of erythropoietin receptor expression in breast cancer. Lab Invest 2002;82:911 ^ 8. 89. Kumar SM, Acs G, Fang D, et al. Functional erythropoietin autocrine loop in melanoma. Am J Pathol 2005;166:823 ^ 30. 90. Acs G, Zhang PJ, McGrath CM, et al. Hypoxiainducible erythropoietin signaling in squamous dysplasia and squamous cell carcinoma of the uterine cervix and its potential role in cervical carcinogenesis and tumor progression. Am J Pathol 2003;162: 1789 ^ 806. 91. Pajonk F, Weil A, Sommer A, Suwinski R, Henke M. The erythropoietin-receptor pathway modulates survival of cancer cells. Oncogene 2004;23: 8987 ^ 91. 92. Belenkov AI, Shenouda G, Rizhevskaya E, et al. Erythropoietin induces cancer cell resistance to ionizing radiation and to cisplatin. Mol Cancer Ther 2004; 3:1525 ^ 32. 93. Carvalho G, Lefaucheur C, Cherbonnier C, et al. Chemosensitization by erythropoietin through inhibition of the NF-nB rescue pathway. Oncogene 2005; 24:737 ^ 45. 94. Mittelman M, Neumann D, Peled A, Kanter P, Haran-Ghera N. Erythropoietin induces tumor regression and antitumor immune responses in murine myeloma models. Proc Natl Acad Sci U S A 2001;98:5181 ^ 6. 95. Tovari J, Gilly R, Raso E, et al. Recombinant human erythropoietin a targets intratumoral blood vessels, improving chemotherapy in human xenograft models. Cancer Res 2005;65:7186 ^ 93. 96. Acs G, Zhang PJ, RebbeckTR, Acs P,Verma A. Immunohistochemical expression of erythropoietin and erythropoietin receptor in breast carcinoma. Cancer 2002;95:969 ^ 81. 97. Yasuda Y, Fujita Y, Masuda S, et al. Erythropoietin is involved in growth and angiogenesis in malignant tumours of female reproductive organs. Carcinogenesis 2002;23:1797 ^ 805. 98. Acs G, Xu X, Chu C, Acs P, Verma A. Prognostic significance of erythropoietin expression in human endometrial carcinoma. Cancer 2004;100:2376 ^ 86. 99. Ribatti D, Marzullo A, Nico B, et al. Erythropoietin as an angiogenic factor in gastric carcinoma. Histopathology 2003;42:246 ^ 50. 100. Arcasoy MO, Amin K, Chou SC, et al. Erythropoietin and erythropoietin receptor expression in head and neck cancer: relationship to tumor hypoxia. Clin Cancer Res 2005;11:20 ^ 7. 101. Dagnon K, Pacary E, Commo F, et al. Expression of erythropoietin and erythropoietin receptor in nonsmall cell lung carcinomas. Clin Cancer Res 2005;11: 993 ^ 9. 102. Arcasoy MO, Amin K,Vollmer RT, et al. Erythropoietin and erythropoietin receptor expression in human prostate cancer. Mod Pathol 2005;18:421 ^ 30. 103. Eccles TG, Patel A, Verma A, et al. Erythropoietin and the erythropoietin receptor are expressed by papillary thyroid carcinoma from children and adolescents. Expression of erythropoietin receptor might be a favorable prognostic indicator. Ann Clin Lab Sci 2003;33:411 ^ 22.

Clin Cancer Res 2006;12(2) January 15, 2006