Pharmacologic treatment options in patients with thrombocytopenia

Pharmacologic treatment options in patients with thrombocytopenia

Pharmacologic Treatment Options in Patients With Thrombocytopenia George D. Demetri Thrombocytopenia that results from chemotherapy has become an incr...

772KB Sizes 0 Downloads 61 Views

Pharmacologic Treatment Options in Patients With Thrombocytopenia George D. Demetri Thrombocytopenia that results from chemotherapy has become an increasingly important issue in the treatment of cancer and remains a difficult clinical problem. The identification of a safe and effective platelet growth factor could significantly improve the management of severe chemotherapy-induced thrombocytopenia. Over the past decade, a number of hematopoietic growth factors with thrombopoietic activity have been identified, including stem-cell factor (c-kit ligand), interleukin (IL)-1, 11-3, IL-6, and IL-11, as well as thrombopoietin (TPO) and its derivatives. Only a few of these agents have shown acceptable tolerability and sufficient ability to stimulate thrombopoiesis to justlfy testing in randomized clinlcal trials. Currently, IL-11 is the only cytokine licensed In the United States for treatment of chemotherapy-induced thrombocytopenia. However, Its thrombopoietic activity is modest and its use is often associated with unfavorable side effects. Identification of TPO, the c-Mpl ligand, as the primary physiologic regulator of megakatyocyte and platelet development offers important promise for treatment of thrombocytopenia. Preliminary clinical studies of recombinant human TPO (rhTPO), a full-length glycosylated molecule, indicate that it is safe and biologically active in reducing severe chemotherapy-induced thrombocytopenia. In addition to rhTP0, the future may see the development of novel genetically englneered, high-afflnity cytokine receptor agonists and c-Mpl ligand mimetic peptides. Semin Hematol37(suppl4):ll-18. Copyright 0 2000 by W.B. Saunders Company.

M

YELOSUPPRESSION AND ASSOCIATED anemia, neutropenia, and thrombocytopenia are common dose-limiting and clinically problematic sequelae of radiation therapy and chemotherapy. Although growth factors such as erythropoietin, granulocyte colony-stimulating factor (G-CSF), and granulocyte macrophage colony-stimulating factor (GM-CSF) have improved the management of chemotherapy-induced anemia’s2and neutropenia,3t4 an unmet medical need remains for mitigation of severe chemotherapy-induced thrombocytopenia. Although thrombocytopenia associated with conventional dose chemotherapy regimens rarely requires intervention, it remains a serious clinical problem in patients undergoing dose-intensive chemotherapy, induction and consolidation therapy of hematologic malignancies, and chemotherapy following multiple prior regimens. Platelet transfusion therapy is currently the only acute treatment for severe thrombocytopenia. Although temporarily effective in controlling severethrombocytopenia, platelet transfusion therapy is associatedwith several problems, including refractoriness and alloimmunization,>-a transmission of infectious agents,9T10 and transfusion reactions.ll The limited supply Seminars

in Hematology,

of blood products and increased cost associated with complications of platelet transfusions can also be problematic. l2 Furthermore, the need for therapeutic platelet transfusions limits outpatient treatment. Consequently, the clinical availability of safe and effective platelet growth factors is eagerly awaited.

Cytokines

with Thrombopoietic Effects

The search for a thrombopoietic growth factor has led to the identification of a number of cytokines and growth factors that can influence thrombopoiesis including thrombopoietin (TPO), stem-cell factor (c-kit ligand), interleukin (IL)-1, IL-3, IL-6, IL-l 1, and GM-CSF (Table 1). Preclinical studies of IL-l, IL-3, IL-6, and IL-l 1 indicated that these cytokines could stimulate megakaryocyte growth and producFrom the Center for Sarcoma and Bone Oncology, Farber Cancer Institute, Sarcoma Center, Boston, MA. Address reprint requests to George D. Demetri, MD,

DanaCenter

for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Sarcoma Center, 44 Binney St, Boston, MA, 02115-6084. Copyright 0 2000 by W.B. Saunders Company 003 7-l 963/00/3 702-4004$10.00/0 doi:lO.lO~3/sh.2000.7388

Vol 37, No 2, SuppI 4 (Apd),

2000:

pp 1 l-18

11

12

George D. Demtri

Table

1. Cytokine

Approaches

to Thrombopoiesis

Pleiotropic cytokines IL-1 IL-3 IL-6 IL-11 c-Mpl llgands Peg-rHuMGDF rhTP0 Abbreviations: IL, interleukin; cyte growth and development human thrombopoietln.

Peg-rHuMGDF, factor: rhTP0,

megakarye recombinant

tion both in vitro and in animal models of severe radiation- and chemotherapy-induced thrombocytopenia. However, the thrombopoietic activity observed in vitro often has not translated into a significant clinical effect, and many of the first thrombopoietic cytokines tested had unacceptable toxicities.

Initial Thrombopoietic

Cytokines

In phase I/II studies, IL-3 functioned as a multilineage hematopoietic growth factor and produced dose-dependent effects on peripheral platelet counts. i3-16 However, these promising results were not confirmed in phase III studies, which showed no significant clinical benefit from adding IL-3 to supportive care in reducing the number of platelet transfusions required or incidence of severe grade 4 thrombocytopenia.17x1sMoreover, IL-3 appeared to have little effect on overall hematopoietic cell recovery in patients with lymphoma undergoing autologous bone marrow transplantation19 and in patients with myelodysplastic syndrome.*O Similarly, variable clinical effect on platelet recovery was observed with stem-cell factor. In a small phase I study of patients with nonsmall-cell lung cancer, stem-cell factor appeared to enhance recovery of neutrophils and platelets after chemotherapy.*’ However, stemcell factor appeared to have no effect on platelet counts in breast cancer patients receiving more dose-intensive therapy.** The pleiotropic biologic effects of the thrombopoietic cytokines also raised concern about their clinical application. Although interleukins such as IL-l and IL-6 have thrombopoietic

activity in vitro, *K** they are associated with a spectrum of in vivo effects, including fever25 and induction of acute-phase proteins.26-29 In clinical studies, the potential therapeutic role of IL- 1(Y in attenuating chemotherapy-induced thrombocytopenia was limited by toxicities, including fever, chills, hypotension, and headache.30J1 Extensive toxiciries observed in patients with solid tumors3* and little efficacy in patients with bone marrow failure also limited the use of recombinant human (rh)IL-1P.33 Similarly, IL-6 has been shown to stimulate murine megakaryocytopoiesis in vivo by acting on early megakaryocyte differentiation.** Although this cytokine appeared to accelerate platelet recovery following chemotherapy with minimal toxicity in patients with ovarian cancer,3* other clinical trials of IL-6 in cancer patients have reported only modest thrombopoietic efficacy and significant toxicity, including hyperbilirubinemia, rapid induction of anemia, fever, and fatigue.29,35v36Thus, moderate efficacy and high toxicity may limit the clinical use of IL-6.

Interleukin-

11

The ability of rhIL-11 (Neumega; Genetics Institute, Cambridge, MA) to indirectly promote the proliferation and maturation of megakaryocytic cells in vitro3’-39 and to accelerate the recovery of platelets in myelosuppressed animals40~41stimulated interest in this cytokine as a potential thrombopoietic agent. In phase I clinical studies conducted in breast cancer patients, administration of rhIL-11 was associated with dose-dependent increases in mean platelet counts, increased bone marrow megakaryocyte and progenitor cell numbers, and reduced incidence of severe thrombocytopenia compared with expectations.**,*3 Similar results were observed in a randomized study of rhIL-11 in breast cancer patients undergoing dose-intensive chemotherapy.** Toxicities associated with rhIL-11 in these studies included fatigue, myalgia, edema, and cardiovascular events.*3~** The ability of rhIL-11 (25 pg/kg and 50 pg/kg) to prevent the need for platelet transfusions compared with placebo was studied in 93 cancer patients who required platelet transfusions in the chemotherapy cycle imme-

Treatment Option

diately before randomization. In this study, rhIL-11 (50 pg/kg) significantly decreased the requirement for platelet transfusions in 30% (eight of 27) of patients (P < .05).45 Based on these results, rhIL-11 was licensed in the United States for the treatment of severe chemotherapy-induced thrombocytopenia. In the autologous transplant setting, rhIL-11 did not reduce the requirement for platelet transfusions and was associated with mild edema and higher incidence of cardiovascular events.46 Thus, despite its ability to ameliorate thrombocytopenia in a subset of patients treated with conventional dose chemotherapy, its moderate toxicity and indirect effect on megakaryocytopoiesis make rhIL-11 a less than ideal thrombopoietic agent.

Novel Strategies: PIXY32 1, Syntbokine, and Combination Therapy Strategies to improve the specificity and activity of thrombopoietic factors have led to the development of several novel factors (Table 2), including the fusion protein PIXY321 and a highly mutated genetically engineered analog of IL-3 referred to as synthokine. PIXY321 is a bifunctional cytokine produced by the fusion of the cDNAs for GM-CSF and IL-3. Results of nonrandomized phase I/II studies of PIXY321 indicated a potential role for this novel agent in attenuating cumulative chemotherapy-induced myelosuppression47-49and in increasing hematologic recovery after autologous bone marrow transplantation.50J1 However, as with the cytokines mentioned previously, favorable preliminary clinical results observed with PIXY321 in phase I/II studies were not confirmed in larger randomized phase III studies5*z53 Furthermore, the development of neutralizing antibodies to PIXY321 abrogated its hematologic effects after multiple exposures.53 Synthokine, a high-affinity IL-3 receptor ligand, also showed greater activity in stimulat-

Table

2. Synthetic

Cytokine

Approaches

PIXY321 IL-3 synthokine c-Mpl ligand mimetic peptides GW395058 Promegapoietin (thrombopoietin

to Thrombopoiesis

plus IL-3 synthokine)

fw

13

Tbrmbocytopenia

ing hematopoiesis of human bone marrow progenitor cells in vitro than did native IL-3.54,55 In a nonhuman primate model of radiationinduced myelosuppression, synthokine at a dose of 100 pg/kg/d significantly reduced the duration of thrombocytopenia compared with control animals.55 However, clinical evaluation of this agent has been discontinued due to lack of sufficient activity to improve thrombocytopoiesis in patients. The role of combined cytokine treatment after cytotoxic therapy has also been investigated. Results of underpowered phase I/II studies evaluating the combination of IL-3 with G-CSF or GM-CSF suggested that this combination was well tolerated and could produce a clinically significant increase in platelet counts in patients receiving myelosuppressive chemotherapy.5”-58 A recent randomized, doubleblind, phase II study evaluated the efficacy of combining IL-6 with G-CSF compared with placebo plus G-CSF in patients receiving standard chemotherapy for ovarian cancer.59 Patients who received IL-6 (1 pglkg) and G-CSF (5 pg/kg) had higher platelet nadirs in the first cycle than did the placebo group. However, no statistically significant difference in cycle treatment delays, carboplatin dose delivered, number of patients with grade 4 thrombocytopenia, or platelet transfusion was observed between the treatment groups. Again, the pattern emerges of initial encouragement from small feasibility studies being trumped by negative results from appropriately powered, randomized studies-a not uncommon theme in the annals of drug development.

Thrombopoeitin

and c-Mpl

Ligands

The cDNA for the c-Mpl ligand was isolated and cloned in 1994,60-64and subsequent studies showed that the c-Mpl ligand is indeed the long sought after TPO. 63,65,66 In both in vitro and in vivo experimental studies, TPO stimulated increasesin megakaryocyte number, ploidy, and platelet count.62,67Thus, TPO appears to be the primary regulator for both the proliferation and differentiation of most stages of megakaryocytopoiesis, although it may not be a critical factor in the final platelet budding from mature

14

George D. Dtmtri

megakaryocytes .68,69 The major recombinant forms of TPO that have been developed for clinical studies include rhTP0 the full-length, glycosylated molecule (Genentech, Inc, San Francisco, CA) and PEG-rHuMGDF the truncated, pegylated derivative of the molecule (Amgen Inc, Thousand Oaks, CA).

Megakaryocyte Growth and Development Factor PEG-rHuMGDF, the most broadly studied variant of TPO, has produced dose-dependent increases in platelet counts in patients with advanced malignancies70 and significantly attenuated chemotherapy-induced thrombocytopenia.71*72Administration of PEG-rHuMGDF to healthy platelet donors increased platelet counts and apheresis yields. These increases, in turn, were associated with significantly larger absolute platelet increments in transfusion recipients. ‘3 However, PEG-rHuMGDF has been associated with adverse events, most importantly the development of neutralizing antibodies.T4 Because of these side effects, the manufacturer (Amgen Inc) withdrew the product from clinical trials in the United States in September 1998.

Recombinant Human Thrombopoietin In clinical studies, rhTP0 has also produced a dose-dependent increase in platelet counts in patients with sarcomas75and gynecologic malignancies76 who were at risk for developing severe chemotherapy-induced thrombocytopenia. In these studies, treatment with rhTP0 was well tolerated and without serious adverse events. Studies of rhTP0 in the treatment of chemotherapy-induced thrombocytopenia continue. To date, the development of neutralizing antibodies in patients treated with rhTP0 has not been reported, which may be due in part to the intravenous route of administration used for rhTP0 compared with the subcutaneous route used for PEG-rHuMGDF. It is postulated that subcutaneous administration may allow dendritic cells in the skin to process PEGrHuMGDF and increase its antigenicity. Further studies of rhTP0 in the clinical setting are all proceeding with the intravenous route of administration to minimize this risk.

Synthetic c-Mpl Ligands Synthetic peptides that share no homology with TPO can be designed to compete with TPO for binding to the c-Mpl receptor. These molecules include the TPO mimetic peptide GW395058 and promegapoietin (Table 2). GW395058 is a pegylated, peptide TPO receptor agonist under evaluation for the treatment of chemotherapyinduced thrombocytopenia.77 Because it can share epitopes, its potential for immunogenicity is a significant concern. However, recent experimental studies suggest that its potential for the production of neutralizing antibodies is low, although the PEG-rHuMGDF experience reinforces the fact that only clinical data will be able to fully answer this important concern.” Promegapoietin, a genetically engineered chimerit growth factor, has agonist activity at both the IL-3 and c-Mpl receptors.78 However, clinical development of this molecule was discontinued in November 1998, again due to concerns of antibody development when given by the subcutaneous route.

Conclusion Platelet transfusions remain the standard of care for the treatment of patients with life-threatening thrombocytopenia. Platelets, however, are a limited and costly resource, and their use is associated with inconvenience, expense, transmissible infections and antibody development. Consequently, significant medical resources have been expended in the search for cytokines and hemacopoietic growth factors to treat and prevent thrombocytopenia. Many of the thrombopoietic cytokines and growth factors identified have failed to produce significant clinical activity. This may be a result of the complex intrinsic biology of platelet production. Although IL-1 1 is approved for the secondary prophylaxis of chemotherapy-induced thrombocytopenia, it is a pleiotropic factor with modest clinical activity and is frequently associated with unacceptable systemic toxicities. Thus, there is room for improvement in cytokine support of thrombopoiesis. The ideal factor must have an outstanding safety and tolerability profile and be effective and reliable in increasing platelet counts. rhTP0 appears to be

Trtutmt

Option

the most specific and effective growth factor identified to date for the treatment of therapyinduced thrombocytopenia. Preliminary clinical evidence indicates that rhTP0 may be a promising adjunct to the conventional approach of platelet transfusion therapy and may become the agent of choice for the treatment of chemotherapy-induced thrombocytopenia in cancer patients.

References

Hematopoietic recovery following high-dose combined alkylating-agent chemotherapy and autologous bone marrow support in patients in phase-I clinical trials of colony-stimulating factors: G-CSF, GMCSF, IL-l, IL-2, M-CSF. Ann Hematol 67:267-276, 1993 4. Heil G, Hoelzer D, Sam. MA, et al: A randomized, double-blind, placebo-controlled, phase III study of filgrastim in remission induction and consolidation therapy for adults with de novo acute myeloid leukemia. Blood 90:47 lo-47 18, 1997 5. Novotny VM: Prevention and management of platelet transfusion refractoriness. VOX Sang 76:1-13, PD: Causes of refractoriness Curr Opin Hematol 2:493-498,

10.

11.

KA, Kitchens aureuJ sepsis Am J Med

12.

13.

15.

16.

CS: Case report: from single-donor Sci 307:340-341,

1994 Chiu EK, Yuen KY, Lie AK, et al: A prospective study of symptomatic bacteremia following platelet transfusion and of its management. Transfusion 34: 950-954, 1994 Chambers LA, Kruskall MS, Pacini DG, et al: Febrile

15

reactions after platelet transfusion: The effect of single versus multiple donors. Transfusion 30:219221, 1990 McCullough J: The potential impact of platelet growth factors in transfusion medicine. Curr Opin Hematol 5:386-390, 1998 Kudoh S, Sawa T, Kurihara N, et al: Phase II study of recombinant human interleukin 3 administration following carboplatin and etoposide chemotherapy in small-cell lung cancer patients. SD2 ILE 964 (IL-3) Study. Cancer Chemother Pharmacol 38:S89-S95, 1996 (suppl) Speyer JL, Mandeli J, Hochster H, et al: A phase I trial of cyclosphosphamide and carboplatinum combined with interleukin-3 in women with advancedstage ovarian cancer. Gynecol Oncol S6:387-394, 1995 Tepler I, Elias A, Kalish L, et al: Effect of recombinant human interleukin-3 on haematological recovery from chemotherapy-induced myelosuppression. Br J Haematol 87:678-686, 1994 D’Hondt V, Weynants P, Humblet Y, et al: Dosedependent interleukin-3 stimulation of thrombopoiesis and neutropoiesis in patients with small-cell lung carcinoma before and following chemotherapy:

A placebo-controlled randomized phase Ib study. J Clin Oncol 11:2063-2071, 1993 17. Newland AC: Is interleukin 3 active in anticancer drug-induced thrombocytopenia? Cancer Chemother Pharmacol 38:S83-S88, 1996 (suppl) 18. Hofstra LS, Kristensen GB, Willemse PH, et al: Randomized trial of recombinant human interleu-

19.

to

1995 7. Contreras M: Diagnosis and treatment of patients refractory to platelet transfusions. Blood Rev 12:215221, 1998 8. Engelfriet CP, Reesink HW, Aster RH, et al: Management of alloimmunized, refractory patients in need of platelet transfusions. VOX Sang 73:191-198, 1997 9. Davda RK, Collins Fatal Staphylococctls platelet transfusion.

Tlmmhxytopenia

14.

1. Platanias LC, Miller CB, Mick R, et al: Treatment of chemotherapy-induced anemia with recombinant human erythropoietin in cancer patients. J Clin Oncol 9:2021-2026, 1991 2. Oberhoff C, Neri B, Amadori D, et al: Recombinant human erythropoietin in the treatment of chemotherapy-induced anemia and prevention of transfusion requirement associated with solid tumors: A randomized, controlled study. Ann Oncol 9:255260, 1998 3. Laughlin MJ, Kirkpatrick G, Sabiston N, et al:

1999 6. Friedberg RC, Mintz platelet transfusion.

fw

20.

21.

22.

23.

kin-3 versus placebo in prevention of bone marrow depression during first-line chemotherapy for ovarian carcinoma. J Clin Oncol 16:3335-3344, 1998 Nemunaitis J, Appelbaum FR, Singer JW, et al: Phase I trial with recombinant human interleukin-3 in patients with lymphoma undergoing autologous bone marrow transplantation. Blood 82:3273-3278, 1993 Nand S, Sosman J, Godwin JE, et al: A phase I/II study of sequential interleukin-3 and granulocytemacrophage colony-stimulating factor in myelodysplastic syndromes. Blood 83:357-360, 1994 Crawford J, Lau D, Erwin R, et al: A phase I trial of recombinant methionyl human stem cell factor (SCF) in patients (pts) with advanced non-small cell lung carcinoma (NSCLC). Proc Am Sot Clin Oncol 12: 135, 1993 (abstr 338) Demetri G, Costa J, Hayes D, et al: A phase I trial of recombinant methionyl human stem cell factor (SCF) in patients with advanced breast carcinoma pre- and post-chemotherapy (CHEMO) with cyclophosphamide (C) and doxorubicin. Proc Am Sot Clin Oncol 12:142, 1993 (abstr) Takahashi T, Tsuyuoka R, Ueda Y, et al: Megakaryocyte potentiating activity of IL-l, IL-6 and GM-CSF

16

Gemge D. Demetri

as evaluated megakaryocytic

by

their colonies.

action on in vitro human Br J Haematol78:480-487,

1991 Hill RJ, Warren MK, Stenberg P, et al: Stimulation of megakaryocytopoiesis in mice by human recombinant interleukin-6. Blood 77~42-48, 1991 Kozak W, Kluger MJ, Soszynski D, et al: IL-6 and IL-1 /3 in fever. Studies using cytokine-deficient (knockout) mice. Ann NY Acad Sci 856:33-47, 1998

38.

Mortensen RF, Shapiro J, Lin BF, et al: Interaction of recombinant IL-1 and recombinant tumor necrosis factor in the induction of mouse acute phase proteins. J Immunol 140:2260-2266, 1988 Zahedi K, et al: IL-1 and IL-6 27. Lin BF, Ku NO, mediate increased production and synthesis by hepatocytes of acute-phase reactant mouse serum amyloid P-component (SAP). Inflammation 14:297-313,

40.

24.

25.

26.

28.

29.

30

31

32.

33.

34.

35.

36.

37.

1990 Banks RE, Forbes MA, Storr M, et al: The acute phase protein response in patients receiving subcutaneous IL-6. Clin Exp Immunol 102:217-223, 1995 Gordon MS, Nemunaitis J, Hoffman R, et al: A phase I trial of recombinant human interleukin-6 in patients with myelodysplastic syndromes and thrombocytopenia. Blood 85:3066-3076, 1995 Smith JWI, Longo DL, Alvord WG, et al: The effects of treatment with interleukin1 a! on platelet recovery after high-dose carboplatin. N Engl J Med 328:756761, 1993 Vadhan-Raj S, Kudelka AP, Garrison L, et al: Effects of interleukin1 (Y on carboplatin-induced thrombocytopema m patients with recurrent ovarian cancer. J Clin Oncol 12:707-714, 1994 Rinehart J, Hersh E, Issell B, et al: Phase 1 trial of recombinant human interleukin-lp (rhIL-lp), carboplatin, and etoposide in patients with solid cancers: Sourhwest Oncology, Group Study 8940. Cancer Invest 15:403-410, 1997 Nemunaitis J, Ross M, Meisenberg B, et al: Phase I study of recombinant human interleukin-l/3 (rhILlp) in patients with bone marrow failure. Bone Marrow Transplant 14:583-588, 1994 D’Hondt V, Humblet Y, Guillaume T, et al: Thrombopoietic effects and toxicity of interleukin-6 in patients with ovarian cancer before and after chemotherapy: A multicentric placebo-controlled, randomized phase Ib study. Blood 85~2347-2353, 1995 Lazarus HM, Winton EF, Williams SF, et al: Phase I multicenter trial of interleukin 6 therapy after autologous bone marrow transplantation in advanced breast cancer. Bone Marrow Transplant 15:935-942, 1995 Nieken J, Mulder NH, Buter J, et al: Recombinant human interleukin-6 induces a rapid and reversible anemia in cancer patients. Blood 86:900-905, 1995 Musashi M, Yang YC, Paul SR, et al: Direct and synergistic effects of interleukin 11 on murine he-

39.

41.

42.

mopoiesis 881765-769, Teramura

in

culture. 1991 M, Kobayashi

Proc

Nat1

S, Hoshino

Acad

Sci

USA

S, et al: Inter-

leukin-1 1 enhances human megakaryocytopoiesis in vitro. Blood 79~327-331, 1992 Weich NS, Wang A, Fitzgerald M, et al: Recombinant human interleukin-11 directly promotes megakaryocytopoiesis in vitro. Blood 90:3893-3902, 1997 Leonard JP, Quint0 CM, Kozitza MK, et al: Recombinant human interleukin-1 1 stimulates mulrilineage hematopoietic recovery in mice after a myelosuppressive regimen of sublethal irradiation and carboplatin. Blood 83:1499-1506, 1994 Maze R, Moritz T, Williams DA: Increased survival and multilineage hematopoietic protection from delayed and severe myelosuppressive effects of a nitrosourea with recombinant interleukin-1 1. Cancer Res 54:4947-4951, 1994 Orazi A, Cooper RJ, Tong J, et al: Effects of recombinant human interleukin-1 1 (Neumega rhIL-11 growth factor) on megakaryocytopoiesis in human bone marrow. Exp Hematol 24:1289-1297,

43.

1996 Gordon MS, McCaskill-Stevens WJ, Battiato LA, et al: A phase I trial of recombinant human interleukin-l 1 (Neumega rhIL-11 growth factor) in women

44.

with breast cancer receiving chemotherapy. Blood 87:3615-3624, 1996 Isaacs C, Robert NJ, Bailey FA, et al: Randomized placebo-controlled study of recombinant human interleukin-1 1 to prevent chemotherapy-induced

45.

thrombocytopenia in patients with breast cancer receiving dose-intensive cyclophosphamide and doxorubicin. J Clin Oncol 15:3368-3377, 1997 Tepler I, Elias L, Smith JW II, et al: A randomized placebo-controlled trial of recombinant human interleukin-1 1 in cancer patients with severe thrombocytopenia due to chemotherapy. Blood 87:3607-3614,

46.

47.

48.

1996 Vredenburgh JJ, Hussein A, Fisher D, et al: A randomized trial of recombinant human interleukin-l 1 following autologous bone marrow transplantation with peripheral blood progenitor cell support in patients with breast cancer. Biol Blood Marrow Transplant 4:134-141, 1998 Vadhan-Raj S, Papadopoulos NE, Burgess MA, et al: Effects of PIXY32 1, a granulocyte-macrophage colony-stimulating factor/interleukin-3 fusion protein, on chemotherapy-induced multilineage myelosuppression in patients with sarcoma. J Clin Oncol 12:715-724, 1994 Vadhan-Raj S, Broxmeyer HE, Andreeff M, et al: In vivo biologic effects of PIXY32 1, a synthetic hybrid protein of recombinant human granulocyte-macrophage colony-stimulating factor and interleukin-3 in

Treatment Options fm Thombqtopenia

cancer

patients

with

normal

hematopoiesis:

A phase I

study. Blood 86:2098-2105, 1995 49. Runowicz CD, Mandeli J, Speyer JL, et al: Phase I/II study of PIXY321 in combination with cyclophosphamide and carboplatin in the treatment of ovarian cancer. Am J Obstet Gynecol 174:1151-1159, 1996 50. Vose JM, Anderson JE, Bierman PJ, et al: Phase I/II trial of PIXY321 to enhance engraftment following autologous bone marrow transplantation for lymphoid malignancy. J Clin Oncol 14:520-526, 1996 5 1. Bishop MR, Jackson JD, O’Kane-Murphy B, et al: Phase I trial of recombinant fusion protein PIXY321 for mobilization of peripheral-blood cells. J Clin Oncol 14:2521-2526, 1996 52. Vose JM, Pandite AN, Beveridge RA, et al: Granulocyte-macrophage colony-stimulating factoriinterleukin-3 fusion protein versus granulocyte-macrophage colony-stimulating factor after autologous bone

53.

54.

55.

56.

57.

58.

59.

marrow transplantation for non-Hodgkin’s lymphoma: Results of a randomized double-blind trial. J Clin Oncol 15:1617-1623, 1997 Miller LL, Korn EL, Stevens DS, et al: Abrogation of the hematological and biological activities of the interleukin-3/granulocyte-macrophage colony-stimulating factor fusion protein PIXY321 by neutralizing anti-PIXY321 antibodies in cancer patients receiving high-dose carboplatin. Blood 93:3250-3258, 1999 MacVittie TJ, Farese AM, Herodin F, et al: Combination therapy for radiation-induced bone marrow aplasia in nonhuman primates using synthokine SC-55494 and recombinant human granulocyte colony-stimulating factor. Blood 87:4129-4135, 1996 Farese AM, Herodin F, McKearn JP, et al: Acceleration of hematopoietic reconstitution with a synthetic cytokine (SC-55494) after radiation-induced bone marrow aplasia. Blood 87:581-591, 1996 Farber L, Haus U, Fuchsel G, et al: Treatment of prolonged chemotherapy induced severe thrombocytopenia with recombinant human interleukin-3-A report on four cases. Anticancer Drugs 8:288-292, 1997 Veldhuis GJ, Willemse PH, Beijnen JH, et al: Paclitaxel, ifosfamide and cisplatin with granulocyte colony-stimulating factor or recombinant human interleukin 3 and granulocyte colony-stimulating factor in ovarian cancer: A feasibility study. Br J Cancer 75:703-709, 1997 Bernstein SH, Fay JP, Christiansen NP, et al: Sequential interleukin-3 and granulocyte-colony stimulating factor prior to and following high-dose etoposide and cyclophosphamide: A phase I/II trial. Clin Cancer Res 3:1519-1526, 1997 Hochster H, Speyer JL, Mandeli JP, et al: A phase II double-blind randomized study of the simultaneous administration of recombinant human interleukin-6 and recombinant human granulocyte colony-stimu-

60.

61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

17

lating factor following paclitaxel and carboplatin chemotherapy in patients with advanced epithelial ovarian cancer. Gynecol Oncol 72:292-297, 1999 de Sauvage FJ, Hass PE, Spencer SD, et al: Stimulation of megakaryocytopoiesis and thrombopoiesis by the c-Mpl ligand. Nature 369:533-538, 1994 Lok S, Kaushansky K, Holly RD, et al: Cloning and expression of murine thrombopoietin cDNA and stimulation of platelet production in viva. Nature 369:565-568, 1994 Kuter DJ, Beeler DL, Rosenberg RD: The purification of megapoietin: A physiological regulator of megakaryocyte growth and platelet production. Proc Nat1 Acad Sci USA 91:11104-11108, 1994 Wendling F, Maraskovsky E, Debili N, et al: cMp1 ligand is a humoral regulator of megakaryocytopoiesis. Nature 369:571-574, 1994 Bartley ‘ID, Bogenberger J, Hunt P, et al: Identification and cloning of a megakaryocyte growth and development factor that is a ligand for the cytokine receptor Mpl. Cell 77:1117-1124, 1994 Gurney AL, Carver-Moore K, de Sauvage FJ, et al: Thrombocytopenia in c-mpl-deficient mice. Science 265:1445-1447, 1994 Kato T, Ogami K, Shimada Y, et al: Purification and characterization of thrombopoietin. J Biochem (Tokyo) 118:229-236, 1995 (erratum, 119:208, 1996) Kaushansky K, Lok S, Holly RD, et al: Promotion of megakaryocyte progenitor expansion and differentiation by the c-Mpl ligand thrombopoietin. Nature 369:568-571, 1994 de Sauvage FJ, Carver-Moore K, Luoh SM, et al: Physiological regulation of early and late stages of megakaryocytopoiesis by thrombopoietin. J Exp Med 183:651-656, 1996 Choi ES, Hokom MM, Chen JL, et al: The role of megakaryocyte growth and development factor in terminal stages of thrombopoiesis. Br J Haematol 95~227-233, 1996 Basser RL, Rasko JE, Clarke K, et al: Thrombopoietic effects of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuPEG-RHUMGDF) in patients with advanced cancer. Lancet 348:1279-1281, 1996 Fanucchi M, Glaspy J, Crawford J, et al: Effects of polyethylene glycol-conjugated recombinant human megakaryocyte growth and development factor on platelet counts after chemotherapy for lung cancer. N Engl J Med 336:404-409, 1997 Moskowitz C, Nimer S, Gabrilove J, et al: A randomized, double-blind, placebo-controlled, dose finding, efficacy and safety study of PEG-rHuPEGRHUMGDF (M) in non-Hodgkin’s lymphoma (NHL) patients (pts) treated with ice (ifosfamide, carboplatin and etoposide). Proc Am Sot Clin Oncol 17:76a, 1998 (abstr 295) Kuter D, McCullough J, Romo J, et al: Treatment

18

74.

75.

George D. Demetri

of platelet (PLT) donors with pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuPEG-RHUMGDF) increases circulating PLT counts (CTS) and PLT aphersis yields and increases platelet increments in recipients of PLT transfusions. Blood 90:579a, 1997 (suppl 1, abstr 2579) Crawford J, Glaspy J, Belani C, et al: A randomized, placebo-controlled, blinded, dose scheduling trial of pegylated recombinant human megakaryocyte growth and development factor (PEG-RHUPEGRHUMGDF) with filgrastim support in non-small cell lung cancer (NSCLC) patients treated with paclitaxel and carboplatin during multiple cycles of chemotherapy. Proc Am Sot Clin Oncol 17:73a, 1998 (abstr 285) Vadhan-Raj S, Murray LJ, Bueso-Ramos C, et al: Stimulation of megakaryocyte and platelet produc-

76.

77.

78.

tion by a single dose of recombinant human thrombopoietin in patients with cancer. Ann Intern Med 126:673-681, 1997 Vadhan-Raj S, Verschraegen C, McGarry L, et al: Recombinant human thrombopoietin (rhTP0) attenuates high-dose carboplatin (C)-induced thrombocytopenia in patients with gynecologic malignancy. Blood 90:580a, 1997 (suppl 1, abstr 2581) de Serres M, Ellis B, Dillberger JE, et al: Immunogenicity of thrombopoietin mimetic peptide GW395058 in BALB/c mice and New Zealand white rabbits: evaluation of the potential for thrombopoietin neutralizing antibody production in man. Stem Cells (Dayt) 17:203-209, 1999 Lefebvre P, Winter JN, Kahn LE, et al: Megakaryocyte ex vivo expansion potential of three hematopoietic sources in serum and serum-free medium. J Hematother 8:199-208, 1999