You are on page 1of 12

The

Oncologist ® This material is protected by U.S. Copyright law.


Unauthorized reproduction is prohibited.
For reprints contact: Reprints@AlphaMedPress.com

New Insights Into Erythropoietin and Epoetin Alfa:


Mechanisms of Action, Target Tissues,
and Clinical Applications
MITCHELL J. WEISS
The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA

Key Words. Erythropoietin · Signal transduction · Epoetin alfa · Anemia · Cancer · Central nervous system · Apoptosis

L EARNING O BJECTIVES
After completing this course, the reader will be able to:
1. Discuss the mechanism of action of endogenous erythropoietin and the therapeutic use of epoetin alfa to stimulate red blood
cell production and improve the quality of life in patients with cancer.
2. Explain how epoetin alfa is being investigated in alternate dosing regimens and for anemia prevention in patients with cancer.
3. Describe how functional endogenous erythropoietin receptor signaling pathways have been demonstrated in numerous non-
erythropoietic tissues, including in the central nervous system, and relate evidence for the roles of erythropoietin and epoetin
alfa beyond erythropoiesis, including the therapeutic implications of these nonerythroid functions.

CME Access and take the CME test online and receive one hour of AMA PRA category 1 credit at CME.TheOncologist.com

A BSTRACT
Recombinant human erythropoietin (epoetin alfa) effects in cultured neurons and in several animal mod-
has proven beneficial for the treatment of various ane- els for neurologic disease. In one clinical trial, epoetin
mias. The mechanism of action of endogenous erythro- alfa appeared to limit functional and histologic damage
poietin and the therapeutic use of epoetin alfa to in patients with stroke. Therefore, in cancer patients
stimulate red blood cell production and improve the receiving chemotherapy, the beneficial effects of epo-
quality of life in cancer patients are reviewed here. etin alfa could be mediated not only through enhanced
Epoetin alfa may also attenuate the cognitive dysfunc- erythrocyte production but also via direct effects on the
tion associated with cancer therapy. Interestingly, nervous system. Further investigation into the non-
functional endogenous erythropoietin receptor signal- erythropoietic effects of epoetin alfa could broaden its
ing pathways have been demonstrated in numerous clinical utility for patients with cancer and also provide
nonerythropoietic tissues. Of particular importance, new therapies for various neurologic disorders. The
epoetin alfa confers neurotrophic and neuroprotective Oncologist 2003;8(suppl 3):18-29

INTRODUCTION acts by binding to its receptor (EPO-R) and subsequently acti-


Erythropoietin (EPO) is an endogenous cytokine that vating intracellular signal transduction pathways [1]. EPO is
is essential for erythrocyte development [1]. In adults, the kid- essential for life: mice with deletions of the EPO gene or the
neys produce and release EPO in response to hypoxia [2]. EPO EPO-R die of anemia in utero [3, 4]. Recombinant human

Correspondence: Mitchell J. Weiss, M.D., The Children’s Hospital of Philadelphia, 316B Abramson Research Building,
Philadelphia, Pennsylvania 19104, USA. Telephone: 215-590-0565; Fax: 215-590-4834; e-mail: weissmi@email.chop.edu
Received August 29, 2003; accepted for publication October 3, 2003. ©AlphaMed Press 1083-7159/2003/$12.00/0

The Oncologist 2003;8(suppl 3):18-29 www.TheOncologist.com


Weiss 19

EPO (epoetin alfa) is used in clinical practice to reduce to the nucleus to activate numerous target genes [1], including
transfusion requirements during surgery [5] and to treat ane- the apoptosis inhibitor Bcl-x [14].
mia of various etiologies, including anemia of chronic kid- The inhibition of apoptosis by the EPO-activated
ney disease [6], cancer-related or cancer treatment-related JAK2/STAT5/Bcl-x pathway (Fig. 1) is important for ery-
anemia [7], anemia related to zidovudine therapy in HIV- throid differentiation. JAK2 deficiency causes embryonic
infected patients [8], and anemia related to ribavirin therapy death due to the absence of definitive erythropoiesis [15].
for hepatitis C virus infection [9]. Furthermore, mice deficient in STAT5a/5b have anemia that
The mechanism of action of EPO in erythropoiesis and correlates with the decreased expression of Bcl-x and
the effects of epoetin alfa in cancer patients are reviewed here. increased apoptosis in early erythroblasts [16]. Finally, full
Subsequently, evidence for the roles of EPO and epoetin alfa Bcl-x knockout mice died in embryogenesis with extensive
outside erythropoiesis and potential therapeutic uses of epo- apoptosis of immature hematopoietic cells [17], and condi-
etin alfa beyond anemia treatment are explored. This review tional hematopoietic-specific Bcl-x knockout mice had
is based on an educational session presented at the American severe anemia [18]. In both models, Bcl-x was required for
Society of Hematology (ASH) meeting held in December the survival of erythroid cells during terminal maturation
2002, updated and expanded as necessary. Accordingly, [18, 19]. A recent study also demonstrated that enforced
many abstracts on EPO biology that were presented at the Bcl-x expression can rescue maturation of EPO-deprived
meeting are discussed herein. While these studies have not yet erythroid progenitors in vitro, suggesting that the major ery-
been published in full in peer-reviewed journals, they reflect thropoietic function of EPO is to prevent apoptosis and that
the current research in this therapeutic area. Bcl-x is a critical effector gene [20]. However, it is impor-
tant to note that EPO and EPO-R null mice exhibit a more
MECHANISM OF ACTION OF EPO IN ERYTHROPOIESIS severe erythropoietic defect than that seen in Bcl-x null ani-
In adult kidneys, hypoxia gives rise to increased EPO mals, indicating that EPO fosters erythropoiesis through
expression stimulated by the DNA binding protein, hypoxia- additional effectors. In addition, the erythroid defect in the
induced factor-1 (HIF-1) [2, 10]. EPO is secreted into the absence of STAT5a/5b is milder than that produced by loss
plasma and, upon arrival in the bone marrow, binds to EPO-Rs of Bcl-x, indicating that STAT5-independent mechanisms
on the surface of erythroid progenitor cells [11]. This asso- for Bcl-x induction exist.
ciation triggers a conformational change that brings EPO-R- In addition to STAT5, EPO induces JAK2-mediated tyro-
associated Janus family tyrosine protein kinase 2 (JAK2) sine phosphorylation and activation of several other intracel-
molecules into close proximity, stimulating their activation lular proteins [1]. Examples include: Shc [21], which, in turn,
by transphosphorylation [1, 12, 13]. Subsequently, JAK2 mol- may activate the signaling pathway involved in erythroid cell
ecules phosphorylate eight tyrosine residues in the cytoplasmic proliferation [22]; phosphatidylinositol 3-kinase (PI3K) [23],
domain of the EPO-R, which then
serve as docking sites for various
Epo
Src homology 2-domain-containing
EpoR P SHC
Grb2
intracellular signaling proteins [1]. RAS
P Jak2 GAB1 SOS
These proteins, in turn, are tyrosine P
P P Vav
phosphorylated and activated. One P SHP1 P
GAP Ca2+
of these proteins is a signal trans- P
P Giα2
SHP2 MAPKKK RAF-1 P
ducer and activator of transcription P P
IRS-2 MAPK
(STAT5) that, on phosphorylation
P
by JAK2, dissociates from the EPO- P STAT5
R, dimerizes, and then translocates PI 3-K
P P
Bcl-x
p70S6K STAT5
P activation Ca2+
Figure 1. JAK2/STAT5/Bcl-x signal AKT DNA Ca2+, Mg2+ - Depend,
transduction pathway of the EPO-R. OSM Endonuclease
PKC
Adapted with permission from Cheung Giα2 c-myb, c-myc,
TF P
CaM kinase
and Miller [1]. Molecular mechanisms c-fos, c-Jun Nuclear NF-κB
of erythropoietin signaling. Nephron Ca2+ GATA-1 EpoR Ca2+ - Calmodulin inhibits E2A SCL
2001;87:215-222. Adapted with per- SCL DNA
mission of S. Karger AG, Medical
and Scientific Publishers, Basel,
Switzerland.
20 EPO: Functions Beyond Erythropoiesis

which may promote survival of erythroid cells [22], and Clinical Practice Guidelines for the Use of Epoetin Alfa in
phospholipase C-γ1, which may play a role in erythroid cell Cancer-Related Anemia
proliferation [24]. In this regard, loss of the p85α subunit of Evidence-based clinical practice guidelines for the use of
PI3K impairs fetal liver erythropoiesis, although this mutation epoetin alfa in patients with cancer have been developed by the
appears to inhibit signaling through the c-Kit receptor to a American Society of Clinical Oncology (ASCO) in conjunc-
greater extent than does mutation of the EPO-R [25]. tion with ASH [38] and by the National Comprehensive
Cancer Network (NCCN) [39]. The ASCO/ASH guidelines
ERYTHROPOIETIC EFFECTS OF EPOETIN ALFA IN provide a thorough overview of the use of epoetin alfa therapy
PATIENTS WITH CANCER in patients with various forms of cancer based on data included
in an evidence report assembled by the Blue Cross Blue Shield
Rationale for Treatment of Cancer-Related Anemia Association Technology Evaluation Center [38]. The panel
Up to 75% of patients with cancer experience anemia dur- reviewed and summarized many clinical studies involving
ing the course of their disease, either due to the cancer itself patients with solid tumors or hematologic malignancies and
(tumor infiltration of the bone marrow and/or anemia of with chemotherapy- or cancer-related anemia. Briefly, the
chronic disease) or to the myelosuppressive effects of ASCO/ASH guidelines recommend the use of epoetin alfa in
chemotherapy or radiation therapy [7, 26]. Several lines of evi- patients with chemotherapy-associated anemia when Hb levels
dence indicate that aggressive treatment of anemia is an impor- decline to ≤10 g/dl, with the decision of whether to treat less
tant aspect of cancer therapy. First, a quantitative review of severe anemia (i.e., Hb of >10 g/dl to <12 g/dl) determined by
published data from patients with cancer suggests that anemia clinical circumstance [38]. Guidelines published by the NCCN
may be an independent prognostic factor for survival, with a also recommend the use of erythropoietic agents for the treat-
65% overall estimated greater relative risk of death [27]. In a ment of cancer-related anemia, but suggest intervention for Hb
retrospective analysis of patients undergoing chemotherapy levels ≤11 g/dl [39]. For anemia associated with hematologic
and thoracic radiotherapy for stage IIIA/IIIB non-small cell malignancies, the ASCO/ASH guidelines support the use of
lung cancer, declining hemoglobin (Hb) levels were highly epoetin alfa therapy in patients with low-grade myelodysplas-
predictive of survival [28]. The 2-year survival rates were 51% tic syndrome [38]. The authors of those guidelines suggest that
for patients whose Hb level decreased <10% from baseline, evidence for epoetin alfa use is less robust in anemic (Hb ≤10
33% when Hb declined by 10%-30%, and 0% when Hb g/dl) patients with multiple myeloma (MM), non-Hodgkin’s
declined by >30% [28]. While these observations might sim- lymphoma (NHL), or chronic lymphocytic leukemia (CLL)
ply reflect impaired erythrocyte production in patients with not receiving chemotherapy, although some significant
more aggressive malignancies or greater systemic illness, there improvements in Hb levels have been reported with epoetin
are some indications that anemia can interfere with cancer alfa treatment [38]. To optimize epoetin alfa therapy for the
therapy. For example, anoxic tumor cells are two to three times treatment of cancer-related anemia, it is important to monitor
more resistant to radiation therapy than are normally oxy- iron status regularly and to treat iron deficiency [40].
genated cells [29, 30]. In addition, low Hb levels have been
associated with tumor hypoxia, lower rates of local failures, Clinical Experience With Epoetin Alfa in the Treatment of
and lower survival rates during radiation therapy for multiple Cancer- and Cancer-Treatment-Related Anemia
tumor types [30-36]. In both community-based studies and randomized,
In addition to the possible negative effects of anemia on placebo-controlled clinical trials, epoetin alfa, 150-300 U/kg
cancer treatment and clinical outcomes, chronic anemia and the or 10,000-20,000 U administered three times weekly (TIW)
resulting fatigue impair the quality of life (QOL) of patients [41-43] or 40,000-60,000 U once weekly (QW) [44-46], has
with cancer, and the use of epoetin alfa to boost erythrocyte been shown to significantly increase Hb levels and reduce
production in those patients with cancer-related anemia has transfusion requirements in anemic patients (generally, Hb
resulted in clinically important and statistically significant ≤11 g/dl) with various solid and hematologic malignancies
improvements in QOL [37]. Although red blood cell transfu- who are undergoing chemotherapy and/or radiation therapy.
sion is also a treatment option for anemia, its associated prob- Overall, approximately two-thirds of patients responded to
lems include the potential for transmission of infections, limited epoetin alfa treatment, experiencing an increase in Hb of 1
availability of adequate blood supplies, and short-term resolu- g/dl after 4 weeks or ≥2 g/dl (or achieving Hb ≥12 g/dl) after
tion of symptoms. Epoetin alfa is more convenient to adminis- 8 weeks of treatment [47]. Epoetin alfa therapy also has been
ter, reduces infectious risks, and maintains more constant shown to significantly increase Hb levels and significantly
steady-state Hb levels. Epoetin alfa also may offer potential improve QOL in anemic (Hb ≤11 g/dl) cancer patients who
benefits beyond stimulating erythropoiesis (described later). are not undergoing chemotherapy [48].
Weiss 21

The effects of epoetin alfa in patients with hematologic demonstrated. For example, a correlation between Hb level
malignancies, including MM, lymphoma, and CLL, have and survival was reported in patients with testicular cancer
been the subjects of more recent investigations. For example, receiving first-line, sequential, high-dose chemotherapy
in a randomized, open-label study in patients with these [54]. Patients whose Hb levels were ≥10.5 g/dl after com-
malignancies and mild anemia (Hb ≥10 g/dl and ≤12 g/dl) pletion of four cycles of chemotherapy had a 3-year overall
undergoing chemotherapy, QW epoetin alfa therapy signifi- survival rate of 87%, compared with 68% for patients
cantly increased Hb levels and significantly improved QOL whose Hb levels were <10.5 g/dl (p < 0.03) [54]. Together,
[49]. In a pilot study, patients with MM or lymphoma these data suggest that further investigation of the effects of
treated with epoetin alfa before high-dose therapy and autol- epoetin alfa on survival is warranted.
ogous peripheral stem cell transplantation required fewer
transfusions after transplantation, and the median number of Early Intervention With Epoetin Alfa for Cancer-Treatment-
units of red blood cells transfused was significantly lower Related Anemia
than that of historic controls [50]. Two recent studies have evaluated the impact of early
intervention with epoetin alfa therapy on patients with breast
Effects of Epoetin Alfa on QOL cancer and baseline Hb levels of ≥9 g/dl and ≤14 g/dl receiv-
Numerous studies have demonstrated that epoetin alfa ing or scheduled to receive adjuvant chemotherapy [55, 56].
improves QOL (such as energy level and ability to perform In both studies, treatment with epoetin alfa, 40,000 U QW for
daily activities) in anemic patients with various tumor types. at least 12 weeks, maintained or improved Hb levels and
In the community-based studies and the randomized, placebo- QOL, whereas decreases in Hb levels and deterioration of
controlled clinical trial discussed above, TIW [41-43] or QW QOL were observed with historical controls (i.e., no epoetin
[44, 46] epoetin alfa therapy significantly improved QOL. alfa therapy) and in patients treated with placebo [55-57].
Moreover, in studies that assessed the impact of disease Similar results were found in an interim analysis of a phase
progression as a confounder of QOL, functional improve- III randomized study of early initiation of epoetin alfa,
ments with epoetin alfa therapy occurred independently of 40,000 U QW, in patients with various tumor types [58].
tumor response [41, 42]. Using data from the randomized, Patients who received epoetin alfa, 40,000 U QW at the start
placebo-controlled trial [43], a multivariate analysis of chemotherapy, maintained higher Hb levels and reported
revealed a positive correlation between increased Hb levels less fatigue than patients who received epoetin alfa once their
and improvements in QOL in patients treated with epoetin Hb levels decreased to <10 g/dl [58]. Together, these data
alfa [51]. Even in patients with mild anemia (Hb ≥10.5 g/dl), suggest that epoetin alfa treatment initiated at the start of
Hb levels increased significantly and were associated with chemotherapy may maintain Hb levels and QOL in patients
both statistically and clinically significant improvements in with cancer. Early intervention with epoetin alfa may be
functional status [37, 43]. In two additional studies that eval- particularly effective during high-dose chemotherapy [59].
uated the effect of epoetin-alfa-associated increases in Hb on
QOL in anemic cancer patients receiving chemotherapy with Alternate Epoetin Alfa Dosing Regimens
or without sequential radiation therapy [52, 53], the greatest Epoetin alfa, 40,000 U QW, has been shown to be clin-
incremental gain in QOL occurred when Hb rose from 11 g/dl ically equivalent to 150 U/kg TIW in healthy subjects [1]. In
to 12 g/dl (range, 11-13 g/dl). These data demonstrate that addition, alternate dosing regimens of epoetin alfa are cur-
correction of anemia, including mild anemia, leads to signifi- rently under investigation to determine the optimal dose and
cant improvements in functional ability and well-being. schedule that will elicit an early and significant increase in
Hb level while improving flexibility of administration. In an
Effects of Epoetin Alfa on Survival of Patients With Cancer interim analysis of an open-label, nonrandomized study in 11
While a positive effect of epoetin alfa on QOL in anemic (Hb ≤11 g/dl) cancer patients receiving chemother-
patients with cancer is firmly established, its effects on apy with or without concomitant or sequential radiation ther-
tumor progression and survival are less clear. Lower mor- apy, epoetin alfa, 60,000 U QW, increased Hb levels by 1.1
tality in patients receiving epoetin alfa was reported in a g/dl at week 4 and by 2.6 g/dl at week 8 [60]. Similar results
randomized, placebo-controlled study [43]. However, the were obtained in an open-label, nonrandomized, mainte-
protocol was not designed or powered to assess this para- nance study of 20 anemic (Hb ≤11 g/dl) cancer patients
meter and did not control for factors that could influence undergoing chemotherapy [61]. Epoetin alfa administered at
survival, such as disease stage, bone marrow involvement, a higher starting dose of 60,000 U QW increased Hb levels
chemotherapy intensity, and disease progression. An asso- by 1.0 g/dl at week 4 and by 2.9 g/dl at week 8, with 86% of
ciation between low Hb level and survival has also been patients achieving increases in Hb of ≥2 g/dl or reaching an
22 EPO: Functions Beyond Erythropoiesis

Hb level of ≥12 g/dl by week 8. In patients who achieved Hb POTENTIAL FUNCTIONS OF EPO IN THE CENTRAL
increases of ≥2 g/dl and then moved into maintenance ther- NERVOUS SYSTEM
apy, epoetin alfa, 120,000 U every 3 weeks, successfully Both EPO and the EPO-R are present in the central ner-
maintained the Hb levels achieved during initial therapy [61]. vous system [65, 76-79], including the retina [80, 81]. Epoetin
These data suggest that epoetin alfa, 60,000 U QW, followed alfa has been shown to actively cross the blood-brain barrier
by maintenance dosing of 120,000 U every 3 weeks is a fea- in animals (although at significantly higher doses than are
sible treatment strategy for cancer patients with mild anemia conventionally used in the treatment of anemia in humans)
who are undergoing chemotherapy. [82] and in human subjects at doses of 40,000 U or 1,500
In summary, epoetin alfa treatment of anemic cancer U/kg administered intravenously [83]. In vitro, EPO protects
patients receiving cytotoxic therapy leads to significant neurons against ischemia-induced glutamate toxicity, the pri-
increases in Hb levels and reductions in transfusion utiliza- mary factor responsible for neuronal cell death related to
tion, effects associated with both statistically and clinically hypoxia [65, 84].
significant improvements in QOL. Additional data suggest a In rodent models, the systemic administration of epo-
potential survival benefit associated with epoetin alfa ther- etin alfa attenuated the extent of concussive brain injury
apy, although it is not known whether this effect could be due and the toxicity of kainate, which is representative of the
to the correction of anemia or whether epoetin alfa may have excitotoxicity found in many forms of brain injury [82].
an intrinsic effect on survival. Administration of epoetin alfa Systemic epoetin alfa also reduced immune damage [82]
by either the TIW or the more convenient QW regimen and inflammation [85] in an experimental model of multi-
results in clinically equivalent effects on Hb, transfusions, ple sclerosis and enhanced neurologic recovery from exper-
and QOL. The availability of darbepoetin alfa, an altered gly- imental spinal cord trauma [79, 86]. Systemic epoetin alfa
cosylated form of erythropoietin with an extended circulating also has been shown to reduce injury from brain ischemia
half-life, provides additional opportunities to increase dosing both in an experimental animal model [87] and in patients
intervals in cancer patients with chemotherapy-related ane- at doses similar to those used in clinical practice [88]. In
mia. The recommended starting dose is 2.25 mcg/kg SC QW addition, epoetin alfa was shown to protect photoreceptors
[62], although less frequent dosing regimens may be possible from light-induced damage [81] and retinal neurons from
[63, 64]. Additional alternate dosing regimens of epoetin alfa acute ischemia and reperfusion injury in rodents [86].
are presently being investigated to further determine their Mouse embryos with null mutations of EPO or the EPO-R
clinical utility. die from severe anemia in midgestation [3, 4] and also exhibit

NONERYTHROID FUNCTIONS OF EPO Table 1. EPO-R expression and potential functions in normal
The EPO-R is expressed in numerous embryonic and adult nonerythroid cells
tissues in humans and mice. Based on these observations, EPO
EPO-R Expression Function
signaling has been suggested to have various nonerythroid
Astrocytes Decreased apoptotic cell death [65]
functions. The EPO-R is present in nonerythroid blood lines
Cardiomyocytes Mitogenic [65, 66]
including myeloid cells, lymphocytes, and megakaryocytes, as
Endothelial cells Mitogenic [65, 67-69]
well as in multiple nonhematopoietic cells, such as endothelial
Endothelin-1 synthesis and release
cells; mesangial, myocardial, and smooth muscle fiber cells; [65, 67, 69]
neural cells; prostate cells, and renal cells (Table 1) [65-73].
Angiogenic response (proliferation and
Moreover, many of these cell types exhibit active EPO signal- migration) [65, 67, 70]
ing pathways and biologic responses. For example, EPO stim- Megakaryocytes Maturation [65, 71]
ulates survival and proliferation of endothelial cells in vitro Mesangial cells Increased proliferation in vitro [65]
and promotes new blood vessel formation in vivo [66, 67, 70]. Myeloid cells Multilineage increase in vitro [65]
In a murine model, EPO stimulated angiogenesis in wound Immunomodulation [65]
healing [74]. In that model, macrophages in granulation tissue
Neurons Trophic effect [65]
formed during wound healing expressed EPO-R, and EPO was
Increased monoamine concentration [65]
shown to stimulate transforming growth factor (TGF)-β1 pro-
Decreased apoptotic cell death [65]
duction by activated macrophages [74]. In mice expressing a
Renal cells Mitogenesis [72]
constitutively active EPO-R, EPO signaling in endothelial
Prostate epithelial cells Mitogenesis [73]
cells was found to function in vascular repair [75]. Some addi-
Vascular smooth Contraction [65]
tional actions of EPO on nonerythroid tissues, in particular the
muscle cells
nervous system, are discussed later.
Weiss 23

increased apoptosis in the brain [90]. However, mice reported excitability [82], a prominent component of many forms of
to express EPO-R exclusively in hematopoietic tissues develop brain injury [100].
normally and exhibit no apparent neurologic deficits [91].
While extended neurologic testing was not reported for these THERAPEUTIC IMPLICATIONS
animals, the data indicate that EPO-R may not be required for The observed neuroprotective and vascular effects of
the development or basal function of the nervous system. EPO and epoetin alfa in various experimental models sug-
Considering the pleiotropic effects of EPO on neuronal tissues gest that epoetin alfa may benefit patients with spinal cord
discussed previously, it is possible that EPO-R signaling plays injuries, stroke, myocardial infarction, multiple sclerosis, or
an adaptive role in limiting the damage incurred from various retinal diseases (macular degeneration, retinitis pigmentosa,
neurologic stresses. If this is the case, then mice expressing the and glaucoma [101]). In one recent study of stroke patients,
EPO-R solely in hematopoietic tissues should be more sensitive a 33,333-U i.v. dose of epoetin alfa, administered within 8
to experimentally induced neurotoxicity. hours after the onset of symptoms and at 24 hours and also
48 hours later, appeared to limit the extent of histologic
Mechanisms of EPO Action in the Central Nervous System damage and improve functional outcomes [88]. Additional
The underlying mechanisms of EPO as a neuroprotec- clinical trials are required to confirm these findings.
tive agent are hypothesized to be multifactorial, with both
direct and indirect beneficial effects on neurons. EPO may Implications for Cancer Therapy
antagonize the cytotoxic effect of glutamate, increase The discovery that nonerythroid tissues respond to
expression of antioxidant enzymes, reduce nitric-oxide- EPO challenges the relatively simple model that its effects
mediated formation of free radicals, normalize cerebral in patients with cancer derive exclusively from enhanced
blood flow, influence neurotransmitter release, and promote red blood cell production. These potential effects of EPO
neoangiogenesis [65]. Hypoxia and injury increase produc- have both positive and negative theoretical implications.
tion of EPO and EPO-R in the brain [78, 92, 93]. Hypoxia For example, epoetin alfa could activate intracellular sig-
also induced EPO expression in the retina through the pro- naling pathways directly in tumors that express EPO-R. In
duction of HIF-1 [81]. Epoetin alfa may mediate neuropro- addition, the ability of EPO to stimulate new blood vessel
tection indirectly by restoring blood flow to the injured formation could enhance perfusion to tumors and support
tissue [94] or act directly on neurons via the activation of their growth or, alternatively, potentiate antitumor thera-
several signaling molecules, which also function in EPO pies by enhancing blood delivery, as previously discussed.
signaling in erythropoiesis. EPO downregulates tyrosine The demonstrated neuroprotective effects of EPO could
phosphatase Src homology 2 domain-containing protein prevent or reduce the neurotoxicities associated with
tyrosine phosphatase and activates the extracellular signal- chemotherapy and radiation therapy. These issues are
regulated kinases ERK1 and ERK2 in cortical neurons, discussed in detail below.
which may enhance the magnitude and duration of signaling
[95]. In rat hippocampal neurons, epoetin alfa was shown to Potential Direct Effects of Epoetin Alfa on EPO-R-Expressing
protect against hypoxia-induced death through activation of Tumors
ERK1 and ERK2 and Akt [96]. In rat cortical neurons, EPO- Because some cancer cells express EPO-R, treatment
mediated protection from excitotoxin- and nitric-oxide- with epoetin alfa could produce a variety of direct effects on
induced apoptosis involved a novel pathway with crosstalk tumors. For example, it has been suggested that, in certain
between the JAK2 and nuclear factor (NF)-κB signaling tumor cell lines and xenografts that express both EPO and
cascades; EPO-R-induced activation of JAK2 led to activa- EPO-R, EPO signaling may promote cancer progression [73,
tion of NF-κB and subsequent increased expression of the 102-104]. In cell lines of colon carcinoma, the Ewing’s sar-
inhibitor-of-apoptosis genes, XIAP and c-IAP2 [97]. In the coma family of tumors, and neuroblastoma, EPO reduced
hippocampus of gerbils, EPO was shown to protect neurons chemotherapy-induced apoptosis through upregulation of
against ischemic injury by upregulating the antiapoptotic antiapoptotic genes [105]. EPO also induced the release of
gene Bcl-x [98]. In addition to inhibition of apoptosis, neu- angiogenic growth factors in cell lines of colon carcinoma, the
roprotection may also occur through reduction of inflamma- Ewing’s sarcoma family of tumors, glioma, and medulloblas-
tion [86, 87, 96], which plays a key role in many forms of toma [105]. In xenografts and blocks of uterine and ovarian
brain injury. NF-κB, which is activated by EPO under tumors, EPO antagonists induced apoptosis and decreased the
oxidative or nitrosative stress [97], is a regulator of inflam- number of blood vessels [106, 107]. Phosphorylation of JAK2
matory genes [99]. Epoetin alfa has further been shown to and STAT5 appeared to be abolished in tumor blocks treated
have a neurotrophic effect [96] and to affect neuronal with EPO antagonists [107].
24 EPO: Functions Beyond Erythropoiesis

Despite these studies, only a few rare case reports have Potential Neuroprotective Effects of Epoetin Alfa in Patients
been published that describe a possible effect of epoetin alfa With Cancer
on cancer progression in humans, and no causal relationship Neuroprotective effects of EPO could benefit cancer
has been identified. One patient with MM reportedly devel- patients, whose neurocognitive function may be affected
oped plasma cell leukemia upon receiving treatment with epo- directly by cancer within the central nervous system or indi-
etin alfa [108]. In another cancer patient treated with epoetin rectly by paraneoplastic effects, toxicities of certain cancer
alfa, rapid growth of a vestibular schwannoma was reported, treatments, or coexisting neurologic or psychiatric disor-
but the exact cause was not determined [109]. ders [117]. Recently published studies suggest that cogni-
Antitumorigenic effects of EPO and epoetin alfa have tive dysfunction is experienced by about 10% of breast
also been reported. Epoetin alfa alone had no effect on cancer survivors who did not receive chemotherapy and by
growth of a lung carcinoma xenograft; however, in synergy approximately 15%-25% of patients with breast cancer
with cisplatin, it induced a fivefold reduction in tumor mass treated with chemotherapy [118-124]. Data from a pilot
[110]. In addition, epoetin alfa treatment, either in the trial in patients with breast cancer receiving adjuvant
absence of chemotherapy or concomitant with mild chemo- chemotherapy indicate that those receiving QW epoetin
therapy for a short duration, produced an antimyeloma effect alfa experienced less cognitive decline than patients receiv-
and prolonged survival in a small study of patients with MM ing placebo [56]. At a 6-month follow-up, both the epoetin
(n = 5) who had advanced disease and an expected survival alfa group and the placebo group exhibited a restoration of
of less than 6 months [111]. Despite a poor prognosis, cognitive function [56]. Although the difference between
patients survived an additional 42-82 months after initiation groups was not statistically significant, improvements were
of epoetin alfa therapy [111]. These findings are consistent generally greater in the epoetin alfa group [56]. The cogni-
with data from a murine model of MM, where tumor regres- tive improvement demonstrated by placebo-treated patients
sion believed to be mediated via activated CD8+ T cells may suggest that a learning response occurred with the
occurred in 50% of mice inoculated with MM cells [112]. In repeated cognitive assessments. Hence, additional studies
addition, severe combined immunodeficient (SCID) mice are required to determine the efficacy of epoetin alfa as a
bearing small subcutaneous ovarian cancer xenografts exhib- neuroprotective agent for use in cancer treatment. In partic-
ited significantly greater tumor regression (p < 0.05) when ular, it is possible that pretreatment with epoetin alfa could
treated with epoetin alfa and cisplatin compared with those limit some of the neurotoxic effects of radiation therapy or
treated with cisplatin alone [113]. Correction of chemother- select chemotherapeutic agents.
apy-induced anemia with epoetin alfa has also been shown to
completely restore the antitumor efficacy of photodynamic CONCLUSIONS
therapy [114] and increase tumor sensitivity to cyclophos- The ability of epoetin alfa to stimulate red blood cell pro-
phamide in rodent models [115]. duction has benefited patients with a variety of anemias,
Overall, data from more than 10 years of epoetin alfa use including those of renal and cancer origin. Just how EPO
in millions of patients indicate that the drug is safe and ben- stimulates erythropoiesis is not fully understood, although one
eficial for use in cancer [116], with no evidence of associated important mechanism is by enhancing erythroid precursor
tumor progression reported in clinical trials. In a randomized, survival by inducing the antiapoptotic molecule Bcl-x. More
placebo-controlled trial, no differences in adverse events recently, studies have illustrated various biologic effects of
were found between anemic cancer patients receiving epo- EPO on nonerythroid tissues, including vascular endothelial
etin alfa and those receiving placebo TIW for up to 28 weeks cells, the nervous system, and selected tumors. These obser-
[43]. The proportions of deaths in that study were compara- vations have clinical implications for patients with cancer as
ble in the epoetin alfa group (14%, 34/251) and the placebo well as neurologic and ischemic disorders. It is likely that fur-
group (18%, 22/124) [43]. Although some patients who died ther research into EPO-R signaling will enhance insight into
had experienced disease progression, none of the deaths were its actions outside the hematopoietic system and provide fur-
considered related to the study medication [43], with the pos- ther opportunities to extend the therapeutic role of epoetin alfa
sible exception of a stroke that occurred in an elderly patient within and beyond the oncology practice.
treated with epoetin alfa who had a history of dyspnea and
paresthesia [43]. Hence, while EPO-R has been shown to ACKNOWLEDGMENT
exist in some tumors and their associated vasculature, the The author thanks Rosemary Mazanet for reviewing the
overall broad experience has been that epoetin alfa does not manuscript.
produce major effects, either positive or negative, on tumor Dr. Weiss is the recipient of a Focused Giving Award
growth or progression in most patients. from Johnson & Johnson and is a consultant for Pfizer.
Weiss 25

R EFERENCES
1 Cheung JY, Miller BA. Molecular mechanisms of erythro- 17 Motoyama N, Wang F, Roth KA et al. Massive cell death of
poietin signaling. Nephron 2001;87:215-222. immature hematopoietic cells and neurons in Bcl-x-deficient
2 Graber SE, Krantz SB. Erythropoietin: biology and clinical mice. Science 1995;267:1506-1510.
use. Hematol Oncol Clin North Am 1989;3:369-400. 18 Wagner KU, Claudio E, Rucker EB 3rd et al. Conditional
3 Wu H, Liu X, Jaenisch R et al. Generation of committed deletion of the Bcl-x gene from erythroid cells results in
erythroid BFU-E and CFU-E progenitors does not require ery- hemolytic anemia and profound splenomegaly. Develop-
thropoietin or the erythropoietin receptor. Cell 1995;83:59-67. ment 2000;127:4949-4958.

4 Lin CS, Lim SK, D’Agati V et al. Differential effects of an 19 Motoyama N, Kimura T, Takahashi T et al. bcl-x prevents
erythropoietin receptor gene disruption on primitive and apoptotic cell death of both primitive and definitive erythro-
definitive erythropoiesis. Genes Dev 1996;10:154-164. cytes at the end of maturation. J Exp Med 1999;189:1691-1698.
5 Faris PM, Ritter MA, Abels RI. The effects of recombinant 20 Dolznig H, Habermann B, Stangl K et al. Apoptosis protection
human erythropoietin on perioperative transfusion require- by the Epo target Bcl-XL allows factor-independent differentia-
ments in patients having a major orthopaedic operation. The tion of primary erythroblasts. Curr Biol 2002;12:1076-1085.
American Erythropoietin Study Group. J Bone Joint Surg
21 He TC, Jiang N, Zhuang H et al. Erythropoietin-induced
Am 1996;78:62-72.
recruitment of Shc via a receptor phosphotyrosine-indepen-
6 National Kidney Foundation. K/DOQI clinical practice dent, Jak2-associated pathway. J Biol Chem 1995;270:11055-
guidelines for anemia of chronic kidney disease, 2002. Am J 11061.
Kidney Dis 2002;39(suppl 1):S1-S266.
22 Koury MJ, Sawyer ST, Brandt SJ. New insights into
7 Rizzo JD, Seidenfeld J, Piper M et al. Erythropoietin: a para- erythropoiesis. Curr Opin Hematol 2002;9:93-100.
digm for the development of practice guidelines. Hematology
(Am Soc Hematol Educ Program) 2001;10-30. 23 Damen JE, Cutler RL, Jiao H et al. Phosphorylation of tyro-
sine 503 in the erythropoietin receptor (EpR) is essential for
8 Saag MS, Levine AM, Leitz GJ et al. Once-weekly epoetin binding the P85 subunit of phosphatidylinositol (PI) 3-
alfa increases hemoglobin and improves quality of life in ane- kinase and for EpR-associated PI 3-kinase activity. J Biol
mic HIV+ patients [abstract]. In: Program and Abstracts of the
Chem 1995;270:23402-23408.
39th Annual Meeting of the Infectious Diseases Society of
America. San Francisco, California 2002:160. 24 Ren HY, Komatsu N, Shimizu R et al. Erythropoietin induces
tyrosine phosphorylation and activation of phospholipase C-
9 Sulkowski M, Wasserman R, Brau N et al. Once-weekly
gamma 1 in a human erythropoietin-dependent cell line. J Biol
recombinant human erythropoietin (epoetin alfa) facilitates
optimal ribavirin (RBV) dosing in hepatitis C virus-infected Chem 1994;269:19633-19638.
patients receiving interferon-α-2b(IFN)/RBV therapy 25 Huddleston H, Tan B, Yang FC et al. Functional p85α gene
[poster]. Presented at the Hepatitis Single Topic Conference is required for normal murine fetal erythropoiesis. Blood
(AASLD). Chicago, Illinois, June 15, 2001. 2003;102:142-145.
10 Wang GL, Semenza GL. General involvement of hypoxia- 26 Groopman JE, Itri LM. Chemotherapy-induced anemia in
inducible factor 1 in transcriptional response to hypoxia. adults: incidence and treatment. J Natl Cancer Inst
Proc Natl Acad Sci USA 1993;90:4304-4308. 1999;91:1616-1634.
11 Broudy VC, Lin N, Brice M et al. Erythropoietin receptor 27 Caro JJ, Salas M, Ward A et al. Anemia as an independent
characteristics on primary human erythroid cells. Blood prognostic factor for survival in patients with cancer: a sys-
1991;77:2583-2590.
tematic, quantitative review. Cancer 2001;91:2214-2221.
12 Remy I, Wilson IA, Michnick SW. Erythropoietin receptor
28 MacRae R, Shyr Y, Johnson D et al. Declining hemoglobin
activation by a ligand-induced conformation change.
during chemoradiotherapy for locally advanced non-small cell
Science 1999;283:990-993.
lung cancer is significant. Radiother Oncol 2002;64:37-40.
13 Livnah O, Stura EA, Middleton SA et al. Crystallographic
evidence for preformed dimers of erythropoietin receptor 29 Teicher BA. Physiologic mechanisms of therapeutic resis-
before ligand activation. Science 1999;283:987-990. tance. Blood flow and hypoxia. Hematol Oncol Clin North
Am 1995;9:475-506.
14 Silva M, Benito A, Sanz C et al. Erythropoietin can induce the
expression of bcl-xL through Stat5 in erythropoietin-dependent 30 Harrison LB, Chadha M, Hill RJ et al. Impact of tumor hypoxia
progenitor cell lines. J Biol Chem 1999;274:22165-22169. and anemia on radiation therapy outcomes. The Oncologist
2002;7:492-508.
15 Parganas E, Wang D, Stravopodis D et al. Jak2 is essential
for signaling through a variety of cytokine receptors. Cell 31 Fyles AW, Milosevic M, Wong R et al. Oxygenation pre-
1998;93:385-395. dicts radiation response and survival in patients with cervix
cancer. Radiother Oncol 1998;48:149-156.
16 Socolovsky M, Nam H, Fleming MD et al. Ineffective ery-
thropoiesis in Stat5a(-/-)5b(-/-) mice due to decreased 32 Girinski T, Pejovic-Lenfant MH, Bourhis J et al. Prognostic
survival of early erythroblasts. Blood 2001;98:3261-3273. value of hemoglobin concentrations and blood transfusions
26 EPO: Functions Beyond Erythropoiesis

in advanced carcinoma of the cervix treated by radiation 45 Sloan JA, Witzig T, Silberstein P et al. Quality of life, blood
therapy: results of a retrospective study of 386 patients. Int J transfusions, and toxicity, in anemic patients with advanced
Radiat Oncol Biol Phys 1989;16:37-42. cancer receiving weekly erythropoietin while on chemotherapy:
results from a phase III randomized double-blind placebo-con-
33 Grogan M, Thomas GM, Melamed I et al. The importance of
trolled study [abstract]. Proc Am Soc Hematol 2002;100:287a.
hemoglobin levels during radiotherapy for carcinoma of the
cervix. Cancer 1999;86:1528-1536. 46 Shasha D, George MJ, Harrison LB. Once-weekly dosing of
epoetin-α increases hemoglobin and improves quality of life
34 Takigawa N, Segawa Y, Okahara M et al. Prognostic factors
in anemic cancer patients receiving radiation therapy either
for patients with advanced non-small cell lung cancer: univari-
concomitantly or sequentially with chemotherapy. Cancer
ate and multivariate analyses including recursive partitioning
2003;98:1072-1079.
and amalgamation. Lung Cancer 1996;15:67-77.
47 Shasha D, George M. Rapid hemoglobin response in anemic
35 Fein DA, Lee WR, Hanlon AL et al. Pretreatment hemoglo-
cancer patients undergoing chemotherapy or chemoradiation
bin level influences local control and survival of T1-T2
therapy receiving once-weekly epoetin alfa treatment [abstract].
squamous cell carcinomas of the glottic larynx. J Clin Oncol
Blood 2002;100:17b.
1995;13:2077-2083.
48 Quirt I, Robeson C, Lau CY et al. Epoetin alfa therapy increases
36 Glaser CM, Millesi W, Kornek GV et al. Impact of hemoglo-
hemoglobin levels and improves quality of life in patients with
bin level and use of recombinant erythropoietin on efficacy of cancer-related anemia who are not receiving chemotherapy and
preoperative chemoradiation therapy for squamous cell carci- patients with anemia who are receiving chemotherapy. J Clin
noma of the oral cavity and oropharynx. Int J Radiat Oncol Oncol 2001;19:4126-4134.
Biol Phys 2001;50:705-715.
49 Straus DJ, Turner RR, Testa MA et al. Epoetin alfa treatment
37 Cella D, Zagari MJ, Vandoros C et al. Epoetin alfa treatment improves quality of life and increases hemoglobin levels during
results in clinically significant improvements in quality of chemotherapy for lymphoma, chronic lymphocytic leukemia
life in anemic cancer patients when referenced to the general (CLL), and multiple myeloma (MM) patients with mild-to-
population. J Clin Oncol 2003;21:366-373. moderate anemia [abstract]. Blood 2002;100:220a-221a.
38 Rizzo JD, Lichtin AE, Woolf SH et al. Use of epoetin in 50 Hunault-Berger M, Lévy V, Dib M et al. Recombinant
patients with cancer: evidence-based clinical practice guidelines human erythropoietin (rHuEpo) before high dose therapy
of the American Society of Clinical Oncology and the (HDT) allows autologous peripheral stem cell transplanta-
American Society of Hematology. Blood 2002;100:2303-2320. tion (APSCT) without red blood cell (RBC) transfusion. A
39 National Comprehensive Cancer Network Clinical Practice pilot study [abstract]. Proc Am Soc Hematol 2002;100:841a.
Guidelines in Oncology. v.1.2003. Cancer and Treatment- 51 Fallowfield L, Gagnon D, Zagari M et al. Multivariate regres-
Related Anemia. Rockledge, PA: National Comprehensive sion analyses of data from a randomised, double-blind,
Cancer Network, Inc., January 2003. placebo-controlled study confirm quality of life benefit of epo-
40 Henry DH. Supplemental iron: a key to optimizing the response etin alfa in patients receiving non-platinum chemotherapy.
of cancer-related anemia to rHuEPO? The Oncologist Epoetin Alfa Study Group. Br J Cancer 2002;87:1341-1353.
1998;3:275-278. 52 Crawford J, Cella D, Cleeland CS et al. Relationship between
41 Demetri GD, Kris M, Wade J et al. Quality-of-life benefit in changes in hemoglobin level and quality of life during
chemotherapy patients treated with epoetin alfa is independent chemotherapy in anemic cancer patients receiving epoetin alfa
of disease response or tumor type: results from a prospective therapy. Cancer 2002;95:888-895.
community oncology study. Procrit Study Group. J Clin Oncol 53 Shasha D, George MJ, Harrison LB. Significant relationship
1998;16:3412-3425. between hemoglobin (Hb) levels and quality of life (QOL) dur-
42 Glaspy J, Bukowski R, Steinberg D et al. Impact of therapy ing chemoradiation: findings from an incremental analysis of a
with epoetin alfa on clinical outcomes in patients with non- 442-patient prospective, community-based epoetin alfa study
myeloid malignancies during cancer chemotherapy in com- [abstract]. Int J Radiat Oncol Biol Phys 2002;54(suppl 1):48.
munity oncology practice. Procrit Study Group. J Clin Oncol 54 Bokemeyer C, Oechsle K, Hartmann JT et al. Treatment-
1997;15:1218-1234. induced anaemia and its potential clinical impact in patients
43 Littlewood TJ, Bajetta E, Nortier JW et al. Effects of epoetin receiving sequential high dose chemotherapy for metastatic
alfa on hematologic parameters and quality of life in cancer testicular cancer. Br J Cancer 2002;87:1066-1071.
patients receiving nonplatinum chemotherapy: results of a ran- 55 Hudis C, Williams D, Gralow JR et al. Epoetin alfa main-
domized, double-blind, placebo-controlled trial. J Clin Oncol tains hemoglobin and quality of life in breast cancer patients
2001;19:2865-2874. receiving conventional adjuvant chemotherapy: final report
[abstract]. PROCRIT Study Group. Proc Am Soc Clin Oncol
44 Gabrilove JL, Cleeland CS, Livingston RB et al. Clinical
2003;22:767.
evaluation of once-weekly dosing of epoetin alfa in
chemotherapy patients: improvements in hemoglobin and 56 O’Shaughnessy J, Vukelja S, Savin M et al. Impact of epoetin
quality of life are similar to three-times-weekly dosing. J Clin alfa on cognitive function, asthenia, and quality of life in
Oncol 2001;19:2875-2882. women with breast cancer receiving adjuvant or neoadjuvant
Weiss 27

chemotherapy. Analysis of 6-month follow-up data 72 Westenfelder C, Biddle DL, Baranowski RL. Human, rat,
[abstract]. Breast Cancer Res Treat 2002;76(suppl 1):S138. and mouse kidney cells express functional erythropoietin
57 Lawless GD, Ford JM. Cumulative prevalence of anemia in receptors. Kidney Int 1999;55:808-820.
early-stage breast cancer (ESBC) patients [abstract]. Blood 73 Feldman L, Wang Y, Rhim JS et al. Human prostate epithe-
2000;96:390b. lial and prostate cancer cells express both erythropoietin and
58 Richart JM, Petruska PJ, Klebert KS et al. A phase III trial functional erythropoietin receptors [abstract]. Proc Am Soc
of epoetin-alfa: early vs late administration in patients Hematol 2002;100:171b.
treated with chemotherapy [abstract]. Blood 2002;100:175b. 74 Arcasoy MO, Amin K, Haroon ZA. Erythropoietin (EPO)
59 Henry DH. Epoetin alfa and high-dose chemotherapy. Semin receptor expression in macrophages is associated with EPO-
Oncol 1998;25(suppl 7):54-57. mediated stimulation of wound healing response, angiogenesis
and transforming growth factor β-1 production [abstract]. Proc
60 Chap L, George M, Glaspy JA. Evaluation of epoetin alfa
Am Soc Hematol 2002;100:681a.
(Procrit®) 60,000 U once weekly in anemic cancer patients
receiving chemotherapy [abstract]. Proc Am Soc Clin Oncol 75 Wang H, Pastore Y, Pool L et al. Gain-of-function mutation of
2002;21:264b. human erythropoietin receptor in mice decreases neointimal
61 Patton J, Kuzur M, Liggett W et al. Epoetin alfa 60,000 U formation [abstract]. Blood 2002;100:680a-681a.
once-weekly followed by 120,000 U every three weeks to 76 Marti HH, Wenger RH, Rivas LA et al. Erythropoietin gene
maintain hemoglobin levels in anemic cancer patients expression in human, monkey and murine brain. Eur J Neurosci
receiving chemotherapy: final report [abstract]. Proc Am 1996;8:666-676.
Soc Clin Oncol 2003;22:754. Presented at the 39th Annual
Meeting of the American Society of Clinical Oncology. 77 Juul SE, Anderson DK, Li Y et al. Erythropoietin and erythro-
Chicago, Illinois, May 31-June 3, 2003. poietin receptor in the developing human central nervous
system. Pediatr Res 1998;43:40-49.
62 Aranesp® (darbepoetin alfa) Prescribing Information.
Amgen Inc. Thousand Oaks, CA. December 17, 2002. 78 Sirén AL, Knerlich F, Poser W et al. Erythropoietin and ery-
thropoietin receptor in human ischemic/hypoxic brain. Acta
63 Glaspy JA, Jadeja JS, Justice G et al. A randomized, active-
control, pilot trial of front-loaded dosing regimens of darbe- Neuropathol (Berl) 2001;101:271-276.
poetin-alfa for the treatment of patients with anemia during 79 Celik M, Gokmen N, Erbayraktar S et al. Erythropoietin pre-
chemotherapy for malignant disease. Cancer 2003;97:1312- vents motor neuron apoptosis and neurologic disability in
1320. experimental spinal cord ischemic injury. Proc Natl Acad
64 Kotasek D, Steger G, Faught W et al. Darbepoetin alfa Sci USA 2002;99:2258-2263.
administered every 3 weeks alleviates anaemia in patients 80 Bocker-Meffert S, Rosenstiel P, Rohl C et al. Erythropoietin
with solid tumours receiving chemotherapy; results of a dou- and VEGF promote neural outgrowth from retinal explants in
ble-blind, placebo-controlled, randomized study. Eur J
postnatal rats. Invest Ophthalmol Vis Sci 2002;43:2021-2026.
Cancer 2003;39:2026-2034.
81 Grimm C, Wenzel A, Groszer M et al. HIF-1-induced erythro-
65 Buemi M, Cavallaro E, Floccari F et al. The pleiotropic effects
poietin in the hypoxic retina protects against light-induced
of erythropoietin in the central nervous system. J Neuropathol
Exp Neurol 2003;62:228-236. retinal degeneration. Nat Med 2002;8:718-724.

66 Jaquet K, Krause K, Tawakol-Khodai M et al. Erythropoietin 82 Brines M. What evidence supports use of erythropoietin as a
and VEGF exhibit equal angiogenic potential. Microvasc Res novel neurotherapeutic? Oncology (Huntingt) 2002;16(suppl
2002;64:326-333. 10):79-89.

67 Ribatti D, Presta M, Vacca A et al. Human erythropoietin 83 Xenocostas A, Cheung WK, Farrell FX et al. Recombinant
induces a pro-angiogenic phenotype in cultured endothelial human erythropoietin crosses the blood brain barrier: the
cells and stimulates neovascularization in vivo. Blood pharmacokinetics of rhEPO in the cerebrospinal fluid after
1999;93:2627-2636. intravenous administration [abstract]. Proc Am Soc Clin
Oncol 2003;22:230.
68 Anagnostou A, Liu Z, Steiner M et al. Erythropoietin recep-
tor mRNA expression in human endothelial cells. Proc Natl 84 Morishita E, Masuda S, Nagao M et al. Erythropoietin recep-
Acad Sci USA 1994;91:3974-3978. tor is expressed in rat hippocampal and cerebral cortical neu-
69 Anagnostou A, Lee ES, Kessimian N et al. Erythropoietin rons, and erythropoietin prevents in vitro glutamate-induced
has a mitogenic and positive chemotactic effect on endothelial neuronal death. Neuroscience 1997;76:105-116.
cells. Proc Natl Acad Sci USA 1990;87:5978-5982. 85 Agnello D, Bigini P, Villa P et al. Erythropoietin exerts an anti-
70 Yasuda Y, Masuda S, Chikuma M et al. Estrogen-dependent inflammatory effect on the CNS in a model of experimental
production of erythropoietin in uterus and its implication in autoimmune encephalomyelitis. Brain Res 2002;952:128-134.
uterine angiogenesis. J Biol Chem 1998;273:25381-25387. 86 Gorio A, Gokmen N, Erbayraktar S et al. Recombinant human
71 Fraser JK, Tan AS, Lin FK et al. Expression of specific high- erythropoietin counteracts secondary injury and markedly
affinity binding sites for erythropoietin on rat and mouse enhances neurological recovery from experimental spinal
megakaryocytes. Exp Hematol 1989;17:10-16. cord trauma. Proc Natl Acad Sci USA 2002;99:9450-9455.
28 EPO: Functions Beyond Erythropoiesis

87 Brines ML, Ghezzi P, Keenan S et al. Erythropoietin crosses 103 Arcasoy MO, Amin K, Karayal AF et al. Functional signifi-
the blood-brain barrier to protect against experimental brain cance of erythropoietin receptor expression in breast cancer.
injury. Proc Natl Acad Sci USA 2000;97:10526-10531. Lab Invest 2002;82:911-918.
88 Ehrenreich H, Hasselblatt M, Dembowski C et al. Erythropoietin 104 Westenfelder C, Baranowski RL. Erythropoietin stimulates
therapy for acute stroke is both safe and beneficial. Mol Med proliferation of human renal carcinoma cells. Kidney Int
2002;8:495-505. 2000;58:647-657.

89 Junk AK, Mammis A, Savitz SI et al. Erythropoietin adminis- 105 Batra S, Perelman N, Luck LR et al. Pediatric tumor cells
tration protects retinal neurons from acute ischemia-reperfusion express erythropoietin and a functional erythropoietin recep-
injury. Proc Natl Acad Sci USA 2002;99:10659-10664. tor that promote angiogenesis and tumor cell survival
[abstract]. Proc Am Soc Hematol 2002;100:736a.
90 Yu X, Shacka JJ, Eells JB et al. Erythropoietin receptor sig-
106 Yasuda Y, Musha T, Tanaka H et al. Inhibition of erythro-
nalling is required for normal brain development. Development
poietin signalling destroys xenografts of ovarian and uterine
2002;129:505-516.
cancers in nude mice. Br J Cancer 2001;84:836-843.
91 Suzuki N, Ohneda O, Takahashi S et al. Erythroid-specific 107 Yasuda Y, Fujita Y, Masuda S et al. Erythropoietin is involved
expression of the erythropoietin receptor rescued its null in growth and angiogenesis in malignant tumours of female
mutant mice from lethality. Blood 2002;100:2279-2288. reproductive organs. Carcinogenesis 2002;23:1797-1805.
92 Sadamoto Y, Igase K, Sakanaka M et al. Erythropoietin pre- 108 Olujohungbe A, Handa S, Holmes J. Does erythropoietin
vents place navigation disability and cortical infarction in accelerate malignant transformation in multiple myeloma?
rats with permanent occlusion of the middle cerebral artery. Postgrad Med J 1997;73:163-164.
Biochem Biophys Res Commun 1998;253:26-32.
109 Falcioni M, Taibah A, De Donato G et al. Fast-growing
93 Bernaudin M, Marti HH, Roussel S et al. A potential role for vestibular schwannoma. Skull Base Surg 2000;2:95-100.
erythropoietin in focal permanent cerebral ischemia in mice.
110 Sigounas G, Collins M, Sallah S et al. Erythropoietin
J Cereb Blood Flow Metab 1999;19:643-651.
enhances antineoplastic activity of chemotherapeutic agents
94 Springborg JB, Ma X, Rochat P et al. A single subcutaneous [abstract]. Proc Am Soc Hematol 2002;100:210b.
bolus of erythropoietin normalizes cerebral blood flow 111 Mittelman M, Zeidman A, Fradin Z et al. Erythropoietin has an
autoregulation after subarachnoid haemorrhage in rats. Br J anti-myeloma effect: a clinical observation supported by ani-
Pharmacol 2002;135:823-829. mal studies [abstract]. Proc Am Soc Hematol 2002;100:391b.
95 Renzi MJ, Thirumalai N, Jolliffe LK et al. Erythropoietin 112 Mittelman M, Neumann D, Peled A et al. Erythropoietin
down regulates SHP1 and induces a sustained activation of induces tumor regression and antitumor immune responses
ERK1/ERK2 in primary cortical neurons [abstract]. Proc in murine myeloma models. Proc Natl Acad Sci USA
Am Soc Hematol 2001;100:77a. 2001;98:5181-5186.
96 Sirén AL, Fratelli M, Brines M et al. Erythropoietin prevents 113 Silver DF, Piver MS. Effects of recombinant human ery-
neuronal apoptosis after cerebral ischemia and metabolic thropoietin on the antitumor effect of cisplatin in SCID mice
stress. Proc Natl Acad Sci USA 2001;98:4044-4049. bearing human ovarian cancer: a possible oxygen effect.
Gynecol Oncol 1999;73:280-284.
97 Digicaylioglu M, Lipton SA. Erythropoietin-mediated neu-
roprotection involves cross-talk between Jak2 and NF-κB 114 Golab J, Olszewska D, Mróz P et al. Erythropoietin restores
signalling cascades. Nature 2001;412:641-647. the antitumor effectiveness of photodynamic therapy in mice
with chemotherapy-induced anemia. Clin Cancer Res
98 Wen TC, Sadamoto Y, Tanaka J et al. Erythropoietin pro- 2002;8:1265-1270.
tects neurons against chemical hypoxia and cerebral ischemic
injury by up-regulating Bcl-xL expression. J Neurosci Res 115 Thews O, Kelleher DK, Vaupel P. Erythropoietin restores
the anemia-induced reduction in cyclophosphamide cytotox-
2002;67:795-803.
icity in rat tumors. Cancer Res 2001;61:1358-1361.
99 Yamamoto Y, Gaynor RB. Role of the NF-kB pathway in
116 Itri LM. The use of epoetin alfa in chemotherapy patients: a
the pathogenesis of human disease states. Curr Mol Med
consistent profile of efficacy and safety. Semin Oncol
2001;1:287-296. 2002;29(suppl 8):81-87.
100 Martin LJ, Al-Abdulla NA, Brambrink AM et al. Neuro- 117 Meyers CA. Neurocognitive dysfunction in cancer patients.
degeneration in excitotoxicity, global cerebral ischemia, and Oncology (Huntingt) 2000;14:75-79; discussion 79, 81-82, 85.
target deprivation: a perspective on the contributions of
apoptosis and necrosis. Brain Res Bull 1998;46:281-309. 118 Ahles TA, Saykin AJ, Furstenberg CT et al. Neuropsychologic
impact of standard-dose systemic chemotherapy in long-term
101 Becerra SP, Amaral J. Erythropoietin: an endogenous retinal survivors of breast cancer and lymphoma. J Clin Oncol
survival factor. N Engl J Med 2002;347:1968-1970. 2002;20:485-493.
102 Acs G, Acs P, Beckwith SM et al. Erythropoietin and ery- 119 Brezden CB, Phillips KA, Abdolell M et al. Cognitive function
thropoietin receptor expression in human cancer. Cancer Res in breast cancer patients receiving adjuvant chemotherapy.
2001;61:3561-3565. J Clin Oncol 2000;18:2695-2701.
Weiss 29

120 O’Shaughnessy JA. Effects of epoetin alfa on cognitive func- in perspective. Clin Breast Cancer 2002;3(suppl 3):S100-
tion, mood, asthenia, and quality of life in women with breast S108.
cancer undergoing adjuvant chemotherapy. Clin Breast Cancer
123 van Dam FS, Schagen SB, Muller MJ et al. Impairment of
2002;3(suppl 3):S116-S120.
cognitive function in women receiving adjuvant treatment
121 Schagen SB, van Dam FS, Muller MJ et al. Cognitive for high-risk breast cancer: high-dose versus standard-dose
deficits after postoperative adjuvant chemotherapy for breast chemotherapy. J Natl Cancer Inst 1998;90:210-218.
carcinoma. Cancer 1999;85:640-650.
124 Wieneke MH, Dienst ER. Neuropsychological assessment of
122 Schagen SB, Muller MJ, Boogerd W et al. Cognitive dys- cognitive functioning following chemotherapy for breast
function and chemotherapy: neuropsychological findings cancer. Psychooncology 1995;4:61-66.

You might also like