US Pharm. 2006;31(4)(Oncology suppl):3-10.     

Myelodysplastic syndrome (MDS) is a collection of disorders that is difficult to manage clinically, as the advanced age of patients at diagnosis renders the administration of therapy challenging. Appropriate treatment options for MDS range from supportive care through blood transfusions or colony-stimulating factors to intensive therapy with chemotherapy or allogeneic stem cell transplantation (alloSCT). Currently, therapies in clinical studies, including lenalidomide, azacitidine, and decitabine, appear promising for the treatment of this disease.

MDS is a heterogeneous group of clonal hematologic disorders characterized clinically and morphologically by ineffective hematopoiesis. This process can lead to varying degrees and combinations of anemia, neutropenia, and thrombocytopenia, which may place patients with the disease at risk for infection, bleeding, and dependence on red blood cell transfusions.1 In addition, MDS can progress to acute myeloid leukemia (AML) in approximately one third of patients.2 It is estimated that 15,000 to 20,000 new cases of MDS are diagnosed annually in the United States.3 The median age at diagnosis of the disease is 60 to 75 years. Due to increases in average life expectancy and growing awareness of MDS, incidence of the disease is expected to rise in the next decade.3,4

ETIOLOGY  AND  RISK  FACTORS
MDS is the most common hematologic disease among the elderly and occurs in a greater proportion of men than women.1 Exposure to certain chemicals has been associated with MDS; agents such as benzene have a clear association with MDS, while smoking tobacco is weakly associated with development of the disease.5 Exposure to antineoplastic alkylating agents and ionizing radiation has been shown to have a clear association with MDS, which usually develops four to seven years after initial exposure.5 The majority of cases of MDS (80% to 90%) are idiopathic (de novo). MDS arising from chemotherapy or ionizing radiation is referred to as secondary, or therapy-induced MDS.2,5 This distinction is important since secondary MDS carries a poorer prognosis compared to de novo MDS.6 A small percentage of patients may have genetic factors leading to MDS, also known as familial MDS.5

SIGNS  AND SYMPTOMS
Patients with MDS may present with signs and symptoms of hematopoietic failure, such as infection, bleeding, bruising, petechiae, pallor, progressive fatigue, or dyspnea on exertion.1 Lymphadenopathy and hepatosplenomegaly are infrequent.7 Many patients present without any symptoms, but rather with incidental findings of anemia, thrombocytopenia, leuko­ penia, or a combination of these on routine laboratory evaluations.1

Initial diagnosis of MDS is made by determining the peripheral blood counts and performing a careful microscopic analysis of the peripheral blood cells. Other crucial diagnostic tests include cytochemistry of bone marrow cells, immuno­ phenotyping, cytogenetics, in vitro characteristics of bone marrow, and molecular genetics. 5 Additional causes of abnormal hematopoiesis, such as vitamin B12 or folate deficiency, aplastic anemia, or human immunodeficiency virus infection, should also be ruled out.

CLASSIFICATION
Different systems have been used to classify MDS. Classification based on the French-American-British (FAB) system consists of five subgroups of MDS: (1) refractory anemia (RA); (2) refractory anemia with ringed sideroblasts (RARS); (3) refractory anemia with excess blasts (RAEB); (4) refractory anemia with excess blasts in transformation (RAEB-t); and (5) chronic myelomonocytic leukemia (CMML). Each subgroup is differentiated by the number of ringed sideroblasts (erythroblasts containing cytoplasmic iron granules arranged in a ring around the nucleus), degree of monocytosis, and percentage of myeloblasts (blasts) in the bone marrow and peripheral blood (table 1).8 An increased number of blasts, which are immature blood cells, may be indicative of leukemia.1 According to this classification system, patients were diagnosed with MDS when the blast percentage was less than 30% and with AML when the blast percentage was greater than 30%.8 As MDS was increasingly recognized as a hematologic disorder, it became apparent that not all patients could be easily classified into one of the FAB subgroups.


Using the FAB system as a framework, the World Health Organization (WHO) developed a revised classification system for MDS (table 2).9 Undoubtedly, the most important change in this new system was the lowering of the blast count required for MDS diagnosis from less than 30% to less than 20%. Although the WHO criteria for staging may provide more prognostic information as compared to the FAB system, even the most recent clinical studies continue to incorporate the FAB classification criteria.


STAGING  AND  PROGNOSIS
The International Prognostic Scoring System (IPSS) is the most widely used grading system for assessing prognosis in patients with MDS.10 Multivariate analysis of 816 patients identified cytogenetic abnormalities, percentage of bone marrow blasts, and the number of cytopenias as the most significant predictors of survival and progression to AML (table 3).10 A person's total score in the IPSS system is equal to the sum of the individual scores for bone marrow blasts, karyotype, and cytopenias. Patients are then separated into four risk groups (low, intermediate-1 [INT-1], intermediate-2 [INT-2], or high) based on the total score (table 4). The higher the IPSS score, the worse the prognosis.10 Interestingly, poorer survival times occurred in patients 60 years and older in the low and INT-1 groups but did not differ substantially in the higher risk groups (INT-2 and high).10



TREATMENT
The course of MDS and response to therapy are influenced by disease stage, patient age, and indiv­ idual prognostic factors. Therefore, treatment for MDS must be personalized. In patients with low-risk disease, goals of therapy include resolution of cyto­ penias, delayed progression to AML, and increased quality of life.3 In patients with high-risk disease, the goal of therapy is to eliminate the abnormal clone, thereby prolonging disease-free and overall survival.3 Consequently, appropriate treatment options range from supportive care with blood transfusions or colony-stimulating factors to intensive therapy with chemotherapy or allogeneic stem cell transplantation (alloSCT). Currently, alloSCT is the only curative therapy for MDS.11

The National Comprehensive Cancer Network Practice Guidelines in Oncology for MDS use a patient's IPSS risk category, age, and performance status to categorize treatment options. Treatment options are separated into two groups: low-intensity therapy for low- and INT-1-risk categories and high-intensity therapy for INT-2- and high-risk categories.11 High-intensity therapies, which consist of chemotherapy and alloSCT, are beyond the scope of this review and are discussed extensively in several articles.12-14 Also, as uniform definitions of response criteria do not exist, not all definitions for response are included; these can be found in the original articles. A list of therapies used for the treatment of MDS can be found in Table 5.




Supportive Care
Supportive care is currently the standard of care for the treatment of MDS and may be clinically appropriate for patients with any IPSS score.11 Patients are monitored for cytopenias and their associated adverse effects and are treated with packed red blood cell (PRBC) transfusions for symptomatic anemia, platelet transfusions for severe thrombocytopenia or bleeding, or antibiotics for infections.

Hematopoietic cytokines can be considered for refractory symptomatic anemias.11 Epoetin alfa (Procrit/Amgen) has demonstrated effectiveness in treating anemia in patients with MDS. In an open-label, multicenter, compassionate treatment trial, 100 patients received subcutaneous (SC) epoetin alfa 150 units/kg three times weekly for a minimum of four weeks. This dose could be increased to 300 units/kg three times weekly if patients had no response to the lower dose. At the conclusion of the study, 10 patients (10%) were deemed to have shown response to hematocrit (Hct) (increase in Hct of 6% from baseline with no transfusions for one month). Eighteen patients (18%) were considered to have shown response to transfusion (50% decrease in transfusion requirement during the final 12 weeks of the study).Epoetin alfa therapy was generally well tolerated.15

Epoetin alfa in combination with granulocyte colony-stimulating factor (G-CSF, filgrastim, Neupogen/ Amgen) has also been studied in the MDS population. A randomized phase II trial evaluated the use of daily SC epoetin beta and filgrastim in patients with RA, RARS, or RAEB according to the FAB classification. Patients were randomized to one of two groups. The first group received filgrastim followed by the epoetin beta/filgrastim combination; the second group received epoetin beta followed by the epoetin beta/filgrastim combination. A total of 56 patients were included in the study, with 28 patients randomized to each group. Complete erythroid response was defined as an increase in hemoglobin (Hgb) to at least 11.5 g/dL. Partial erythroid response was defined as in increase in Hgb by 1.5 g/dL in nontransfused anemia and a 100% reduction of transfusion need in combination with stable Hgb for four weeks or more in patients with pretreatment transfusion needs. Eighteen of the 47 evaluated patients (38%) had an erythroid response (CR + PR) to treatment, and 10 patients (21%) demonstrated a complete response. There were no significant differences, including response rates, between the two groups. Reported adverse events were mild and included flu-like symptoms and local injection site irritation.16

Other studies using various forms of epoetin (alfa or beta) and dosing schedules with filgrastim have shown similar improvements in erythroid response.17-19 Epoetin alfa in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF, sargramostim, Leukine/Berlex) has also been studied in the MDS population, although response rates are generally lower.20 The use of G-CSF and GM-CSF alone for the treatment of MDS has resulted in improved neutropenia rates; however, data showing reduced infectious episodes, survival prolongation, or reduced risk of transforming to AML do not exist for these agents.21 Interleukin-11 (Oprelvekin, Neumega/Wyeth) has also been studied in a small number of patients with MDS, although the patients could concomitantly receive epoetin alfa for treatment of anemia and/or G-CSF for treatment of neutropenia during the study.22 In addition, darbepoetin alfa (Aranesp/Amgen) has produced 45% erythroid response rates in a recent study of 48 patients with MDS.23

Low-Intensity Therapies
Azacitidine: Azacitidine (Vidaza/ Pharmion) exerts its antineoplastic effects by causing hypometh­ yl­ ation of DNA and direct cytotoxicity on abnormal hema­ topoietic cells in the bone marrow. It is thought that hypo­ methylation may restore normal function to genes needed for differentiation and proliferation.24 Azacitidine is incorporated into DNA where it shows dose- and time-dependent inhibition of methyltransferase activity.25 Azacitidine is rapidly absorbed after SC administration; approximately 89% of the dose is absorbed based on area under the curve. Mean half-life after SC administration is 41 +/- 8 minutes. Urinary excretion is the primary method of elimination.24

Azacitidine is approved for patients with MDS of all five FAB subtypes--RA or RARS (if accompanied by neutropenia or thrombocytopenia or requiring transfusions), RAEB, RAEB-t, and CMML. The recommended starting dose is 75 mg/m2 SC daily for seven days every four weeks. The dose may be increased to 100 mg/m2 if no beneficial effects are seen after two treatment cycles and if the patient experiences no toxicity other than nausea and vomiting. Other dosing adjustments are required based on hematology laboratory values (tables 6, 7). 24 Azacitidine dose should be reduced by 50% if patients experience an unexplained reduction in serum bicarbonate level of less than 20 mEq/L. Similarly, if the blood urea nitrogen or serum creatinine levels become elevated, the dose of azacitidine should be held until the values return to normal or baseline. The subsequent dose should be reduced by 50% for the next treatment course.24







A pivotal phase III study of the Cancer and Leukemia Group B (CALGB) compared supportive care and SC administration of azacitidine in patients meeting the FAB classification of MDS. Patients with RA or RARS were required to show additional signs of significant marrow dysfunction, such as symptomatic anemia requiring PRBC transfusions for at least three months before study entry, thrombocytopenia with two or more platelet counts <=50 ¥ 109/L or a significant hemorrhage requiring platelet transfusions, or neutropenia with an absolute neutrophil count (ANC) <1 ¥ 109/L and an infection requiring intravenous antibiotics.26

This randomized open-label study enrolled 191 patients, with 99 patients receiving azacitidine and 92 patients receiving supportive care. Azacitidine 75 mg/m2/day was administered SC in seven-day cycles beginning on days 1, 29, 57, and 85. The azacitidine dose could be increased to 100 mg/m2/day according to previously stated criteria. Patients were assessed after the fourth cycle. Definitions of response criteria are included in table 8.26




In the azacitidine group, 60% of patients showed response (P<.0001), 7% had a complete response, 16% showed a partial response, and 37% demonstrated improvement. Of the 92 patients randomized to supportive care, none had a complete or partial response and 5% met the criteria for improvement. Trilineage response was 23% for azacitidine and 0% for supportive care. The median time to AML transformation or death was 21 months in patients who received azacitidine and 12 months in patients who received supportive care (P=.007). Forty-nine patients crossed over from the supportive care group; 47% of those patients showed response, 10% had a complete response, 4% showed a partial response, and 33% demonstrated improvement. The median survival was 20 months in the azacitidine group, compared to 14 months in the supportive care group (P=.10). To reduce the confounding data regarding the crossover patients, a second analysis was performed comparing patients in the azacitidine group to those in the supportive care group who did not cross over or who crossed over late in the study (after six months). In this analysis, azacitidine had improved median survival, compared to the supportive care subgroup (P=.03). The most common toxicity of azacitidine was myelosuppression. Grade 3 or 4 neutropenia occurred in 59%, granulocytopenia in 81%, and thrombocytopenia in 70% of patients who received azacitidine.26 Other common adverse events of azacitidine that have been reported in clinical trials include nausea, vomiting, pyrexia, diarrhea, constipation, injection site erythema, and ecchymosis.24

In a separate evaluation of the previous study, quality-of-life assessments were analyzed. Patients in the azacitidine treatment group experienced greater improvement in fatigue (P =.001), dyspnea (P=.0014), physical functioning (P=.0002), psychological distress (P=.015), and positive affect (P=.0077), compared with patients in the supportive care group.27

Lenalidomide: Lenalidomide (Revlimid/Celgene Corp.) is an immunomodulatory agent with a mechanism of action similar to thalidomide.28 An open-label, single-center trial evaluated the use of lenalidomide in 43 patients with MDS who had symptomatic anemia. Patients were randomized to receive lenalidomide 25 mg daily continuously, 10 mg daily continuously, or 10 mg daily for 21 days in every 28-day cycle. All treatment groups received lenalidomide orally. Sequential dose reductions were permitted. Patients included in the study had a diagnosis of MDS based on the FAB criteria for greater than three months and either symptomatic anemia (Hgb<10 g/dL) or transfusion dependence (>=4 units PRBC in previous eight weeks). Patients also had no response to either recombinant erythropoietin or an endogenous erythropoietin level of >500 mU/mL. Overall, 24 patients (56%) had a hematologic response. In 32 patients who were previously transfusion dependent, 20 achieved transfusion independence. After 81 weeks of evaluation, the median duration of a major response had not been reached. Of the 10 patients who achieved a complete cytogenetic response (absence of pretreatment cytogenetic abnormalities), nine had the del (5q) chromosomal abnormality. In addition, 83% of patients with the chromosomal abnormality del (5q) experienced an erythroid response, compared with 57% of patients with a normal karyotype and 12% of patients with other cytogenetic abnormalities. Neutropenia and thrombocytopenia were the most common adverse events, resulting in treatment interruption or dose reduction in 25 patients (58%). 29 In this study, patients who received lenalidomide did not have high incidences of the dose-limiting toxicities associated with thalidomide (i.e., fatigue, constipation, or shortness of breath).

Due to the results of this study and an abstract presented at the annual American Society of Clinical Oncology (ASCO) meeting in 2005, the FDA approved lenalidomide for the treatment of transfusion-dependent anemia due to low- or INT-1-risk MDS associated with a del (5q) cytogenetic abnormality with or without additional cytogenetic abnormalities. Similar to thalidomide's System for Thalidomide Education and Prescribing Safety (S.T.E.P.S.) program, lenalidomide's RevAssist program requires that prescribers, pharmacists, and patients register with the program to prescribe, dispense, or receive the medication, due to the potential risk of teratogenicity.30,31

Investigational Therapies
DNA Methyltransferase Inhibitors: The cytosine analog decitabine (Dacogen/MGI Pharma), originally synthesized in the 1960s, is an analog of azacitidine capable of inhibiting DNA methyltransferase.32,33 The results of a randomized, open-label, phase III trial enrolling 170 patients were recently reported at the 2005 annual ASCO meeting. The study compared decitabine (n=89) plus supportive care to supportive care alone (n=81) in patients with IPSS INT-1 (31%), INT-2 (44%), and high-risk (26%) MDS. Decitabine was administered as a three-hour infusion of 15 mg/m2 every eight hours for three consecutive days every six weeks. Response rates were 17% for decitabine, compared to 0% for supportive care alone (P<.001). Of the 15 patients (17%) who achieved a response, eight (9%) had complete responses and seven (8%) had partial responses. In addition, hematologic improvements were observed in an additional 13% of patients who received decitabine and 7% of patients who received supportive care. The probability of progression to AML or death was 1.72-fold greater in the supportive care group than in the decitabine group (P=.017). Median time to progression to AML or death was 340 days in patients receiving decitabine, compared to 219 days in patients receiving supportive care (P=.043). Myelosuppression was the most common toxicity in the decitabine group, and febrile neutropenia was the most common grade 3 or 4 toxicity (most serious toxicity, range 0 to 4). 34 Additional data in this study are limited due to the abstract format of the report. These results are the basis of an FDA submission, currently under review for the treatment of MDS.35

Antiangiogenic Therapies: Thalidomide (Thalomid/Celgene Corp.) is probably most widely known for its teratogenicity, which caused a variety of deformities in infants, specifically, amelia (lack of limb) and phocomelia (seal limb).36 Thalidomide is a potent inhibitor of vascular endothelial growth factor and basic fibroblast growth factor, which are both needed for angiogenesis. These angiogenic factors as well as others have been identified in the bone marrow, plasma, and blood cells of patients with MDS.37 Four phase II studies have evaluated thalidomide as single-agent therapy in MDS.

In the largest phase II study to date, patients with MDS of all morphological subtypes received thalidomide at doses ranging from 100 to 400 mg/day given at bedtime. A total of 83 patients were enrolled, with 32 patients discontinuing thalidomide before 12 weeks of treatment. Of these 32 patients, one patient never started thalidomide, six had disease progression, 11 had other medical problems, and 14 discontinued therapy due to side effects. At study end, there were no patients with a complete response. Of the 16 patients with hematologic improvement, 15 patients had an erythroid response and one patient had a platelet response. The most common side effects in this study were fatigue (79%), constipation (71%), and shortness of breath (54%). However, fewer than 5% of patients experienced grade 4 toxicity.38

The major benefit of thalidomide in the treatment of patients with MDS is transfusion independence. Further studies are needed to assess the durability of hematologic response and impact on quality of life.

Farnesyltransferase Inhibitors: The Ras system of proteins is a cellular signaling pathway that controls cell growth, proliferation, and cell death. Experimental studies have shown that mutated forms of Ras have been found in a wide range of malignancies, including MDS.39,40 A key process in the Ras pathway is prenylation, which is carried out by one of two enzymes--farnesyltransferase or geranylgeranyltransferase.39 The most extensively studied farnesyltransferase inhibitor in MDS is tipifarnib (Zarnestra/Johnson & Johnson). A phase II study evaluated the use of tipifarnib in patients with MDS with intermediate- (INT-1, INT-2) or high-risk IPSS scores. Patients were treated with tipifarnib at a starting dose of 600 mg by mouth twice daily for 28 days followed by a two-week rest period (six weeks = one course). Of the 28 patients enrolled, three patients showed response, with two having a complete response and one demonstrating a partial response. The most common side effect was myelosuppression, with 79% of patients experiencing anemia, 75% experiencing thrombo­ cytopenia, and 61% experiencing neutropenia.41 Additionally, 11% and 21% of patients receiving tipifarnib experienced neurotoxicity and rash, respectively.41 In a second phase II study presented at the annual meeting of the American Society of Hematology, 82 patients with MDS were treated with tipifarnib, resulting in responses seen in 28 patients (33%).42

Recently, the FDA's Oncologic Drugs Advisory Committee rejected an accelerated approval request for tipifarnib in patients with AML.43 Studies with tipifarnib in patients with MDS are ongoing. 

In addition to the previously mentioned agents, other novel therapies are undergoing clinical trials. Arsenic trioxide is currently under evaluation in phase II clinical trials in patients with high- and low-risk MDS. Lonafarnib, a farnesyltransferase inhibitor, has undergone phase II trials in patients with advanced MDS and CMML. Small-molecule inhibitors of the VEGF receptor tyrosine kinases SU5416 and SU11248 have produced dose-limiting nonhematologic toxicity in some cases and low response rates. Bevacizumab is currently undergoing phase II trials in MDS. Other agents of interest include bortezomib (proteasome inhibitor), TLK199 (liposomal glutathione derivative), and antisense oligonucleotides to inactivated p53 RNA.21

Conclusion
MDS is a collection of disorders that is difficult to manage clinically. The advanced age of patients at diagnosis makes administration of cytotoxic chemotherapy or alloSCT challenging. Furthermore, with the exception of alloSCT, there are no curative therapies available for treatment of MDS, and supportive care continues to remain the standard of care. Currently, azacitidine and lenalidomide are the only FDA-approved treatments for MDS, although several other therapies are either in clinical trials or the approval process. Some of these investigational therapies currently in clinical studies appear promising, and patients with this uncommon disorder stand to benefit from the availability of additional agents.

REFERENCES
1. Heaney ML, Golde DW. Myelodysplasia. N Engl J Med. 1999;340:1649-1660.
2. Leone G, Mele L, Pulsoni A, et al. The incidence of secondary leukemias. Haematologica. 1999;84:937-945.
3. Rowe JM. State of the science for myelodysplastic syndrome: prognosis and promise of new therapies. Best Pract Res Clin Haematol. 2004;17:535-541.
4. Aul C, Germing U, Gattermann N, Minning H. Increasing incidence of myelodysplastic syndromes: real or fictitious? Leuk Res. 1998;22:93-100.
5. Mufti GJ. Pathobiology, classification, and diagnosis of myelodysplastic syndrome. Best Pract Res Clin Haematol. 2004;17:543-557.
6. Estey EH. Prognosis and therapy of secondary myelodysplastic syndromes. Haematologica . 1998;83:543-549.
7. Faderl S, Kantarjian HM. Novel therapies for myelodysplastic syndromes. Cancer. 2004;101:226-241.
8. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the myelodysplastic syndromes. Br J Haematol. 1982;51:189-199.
9. Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood. 2002;100:2292-2302.
10. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079-2088.
11. National Comprehensive Cancer Network. Practice Guidelines in Oncology: Myelodysplastic Syndromes. Version 1. 2005. Available at: www.nccn.org/ professionals/physician_gls/PDF/mds.pdf. Accessed July 26, 2005.
12. Foss FM. Nucleoside analogs and antimetabolite therapies for myelodysplastic syndrome. Best Pract Res Clin Haematol. 2004;17:573-584.
13. Giralt S. Bone marrow transplant in myelodysplastic syndromes: new technologies, same questions. Curr Hematol Rep. 2005;4:200-207.
14. McCarty J. Transplant strategies for myelodysplastic syndrome. Best Pract Res Clin Haematol. 2004;17:559-572.
15. Rose EH, Abels RI, Nelson RA, et al. The use of r-HuEpo in the treatment of anaemia related to myelodysplasia (MDS). Br J Haematol. 1995;89:831-837.
16. Hellstrom-Lindberg E, Ahlgren T, Beguin Y, et al. Treatment of anemia in myelodysplastic syndromes with granulocyte colony-stimulating factor plus erythropoietin: results from a randomized phase II study and long-term follow-up of 71 patients. Blood. 1998;92:68-75.
17. Negrin RS, Stein R, Vardiman J, et al. Treatment of the anemia of myelodysplastic syndromes using recombinant human granulocyte colony-stimulating factor in combination with erythropoietin. Blood. 1993;82:737-743.
18. Mantovani L, Lentini G, Hentschel B, et al. Treatment of anaemia in myelodysplastic syndromes with prolonged administration of recombinant human granulocyte colony-stimulating factor and erythropoietin. Br J Haematol. 2000;109:367-375.
19. Casadevall N, Durieux P, Dubois S, et al. Health, economic, and quality-of-life effects of erythropoietin and granulocyte colony-stimulating factor for the treatment of myelodysplastic syndromes: a randomized, controlled trial. Blood. 2004;104:321-327.
20. Thompson JA, Gilliland DG, Prchal JT, et al. Effect of recombinant human erythropoietin combined with granulocyte/macrophage colony-stimulating factor in the treatment of patients with myelodysplastic syndrome. Blood. 2000;95:1175-1179.
21. Jabbour EJ, Giles FJ. New agents in myelodysplastic syndromes. Curr Hematol Rep. 2005;4:191-199.
22. Tsimberidou AM, Giles FJ, Khouri I, et al. Low-dose interleukin-11 in patients with bone marrow failure: update of the M.D Anderson Cancer Center experience. Ann Oncol. 2005;16:139-145.
23. Stasi R, Abruzzese E, Lanzetta G, et al. Darbepoetin alfa for the treatment of anemic patients with low- and intermediate-1-risk myelodysplastic syndromes. Ann Oncol. 2005:16;1921-1927.
24. Pharmion Corporation. Vidaza package insert. Boulder, CO. August 2004. Available at: www.vidaza.com/corporateweb/vidazaus/homeb.nsf/AttachmentsBy Title/Packaging/$FILE/FullPrescribingInformationForVidaza.pdf.
25. Silverman LR. DNA methyltransferase inhibitors in myelodysplastic syndrome. Best Pract Res Clin Haematol. 2004;17:585-594.
26. Silverman LR, Demakos EP, Peterson BL, et al. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 2002;20:2429-2440.
27. Kornblith AB, Herndon JE 2nd, Silverman LR, et al. Impact of azacytidine on the quality of life of patients with myelodysplastic syndrome treated in a randomized phase III trial: a Cancer and Leukemia Group B study. J Clin Oncol. 2002;20:2441-2452.
28. Sekeres MA, List A. Lenalidomide (Revlimid, CC-5013) in myelodysplastic syndromes: is it any good? Curr Hematol Rep. 2005;4:182-185.
29. List A, Kurtin S, Roe DJ, et al. Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med. 2005;352:549-557.
30. List AF, Dewald G, Bennett J, et al. Hematologic and Cytogenetic (CTG) Response to Lenalidomide (CC-5013) in Patients with Transfusion-Dependent (TD) Myelodysplastic Syndrome (MDS) and Chromosome 5q31.1 Deletion: Results of the Multicenter MDS-003 Study. Poster presented at 41st Annual American Society of Clinical Oncology Meeting. May 13-17, 2005; Orlando, FL.
31. Celgene Corporation. Revlimid package insert. Summit, NJ . December 2005. Available at: www.celgene.com/PDF/RevlimidPI.pdf.
32. Issa JP. Decitabine. Curr Opin Oncol. 2003;15:446-451.
33. Saba HI, Wijermans PW. Decitabine in myelodysplastic syndromes. Semin Hematol. 2005;42(3 suppl 2):S23-S31.
34. Saba HI, Rosenfeld CS, Issa J, et al. Clinical benefit and survival endpoints from a phase III trial comparing decitabine (DAC) vs. supportive care (SC) in patients with advanced myelodysplastic syndromes (MDS). Poster presented at 41st Annual American Society of Clinical Oncology Meeting. May 13-17, 2005; Orlando, FL.
35. SuperGen, Inc. Web site. Dacogen NDA Accepted for Filing by FDA. Available at: ir.supergen.com/phoenix.zhtml?c=105560&p=irol-newsArticle&ID=658150& highlight=. Accessed August 8, 2005.
36. Matthews SJ, McCoy C. Thalidomide: a review of approved and investigational uses. Clin Ther. 2003;25:342-395.
37. Estey EH. Modulation of angiogenesis in patients with myelodysplastic syndrome. Best Pract Res Clin Haematol. 2004;17:623-639.
38. Raza A, Meyer P, Dutt D, et al. Thalidomide produces transfusion independence in long-standing refractory anemias of patients with myelodysplastic syndromes. Blood. 2001;98:958-965.
39. Feldman EJ. Farnesyltransferase inhibitors in myelodysplastic syndrome. Curr Hematol Rep . 2005;4:186-190.
40. Boguski MS, McCormick F. Proteins regulating Ras and its relatives. Nature. 1993;366:643-654.
41. Kurzrock R, Albitar M, Cortes JE, et al. Phase II study of R115777, a farnesyl transferase inhibitor, in myelodysplastic syndrome. J Clin Oncol. 2004;22:1287-1292.
42. Kurzrock R, Fenaux P, Raza A, et al. High-Risk Myelodysplastic Syndrome (MDS): First results of international phase 2 study with oral farnesyltransferase inhibitor R115777 (Zarnestra). Presented at the 46th Annual Meeting of the American Society of Hematology. December 5, 2004; San Diego, CA .
43. FDA Advisory Committee Web site. Zarnestra Phase III Data Needed Pre-Approval, Oncology Committee Says. Available at: www.fdaadvisorycommittee. com/FDC/AdvisoryCommittee/Committees/Oncologic+Drugs/050505_ zarnestra/050505_ZarnestraR.htm. Accessed August 3, 2005.

To comment on this article, contact editor@uspharmacist.com.