51
|
Thomas D, Vadas M, Lopez A. Regulation of haematopoiesis by growth factors - emerging insights and therapies. Expert Opin Biol Ther 2004; 4:869-79. [PMID: 15174969 DOI: 10.1517/14712598.4.6.869] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Haematopoiesis is regulated by a wide variety of glycoprotein hormones, including stem cell factor, granulocyte-macrophage colony-stimulating factor, thrombopoietin and IL-3. These haematopoietic growth factors (HGFs) share a number of properties, including redundancy, pleiotropy, autocrine and paracrine effects, receptor subunit oligomerisation and similar signal transduction mechanisms, yet each one has a unique spectrum of haematopoietic activity. Ongoing studies with knockout mice have discovered previously unrecognised physiological roles for HGFs, linking haematopoiesis to innate immunity, pulmonary physiology and bone metabolism. The regulation of stem cells by HGFs within niches of the bone marrow microenvironment is now well recognised and similar mechanisms appear to exist in the regulation of other stem cell compartments. Alternative signalling strategies, other than tyrosine kinase activation and phosphotyrosine cascades, may account for some of the more subtle differences between HGFs. Accumulating evidence suggests that some, but not all, HGF receptors can transduce a genuine lineage-determining signal at certain points in haematopoiesis. Further studies, primarily at the receptor level, are needed to determine the mechanisms of instructive signalling, which may include phosphoserine cascades. Novel haematopoietic regulators, as well as the development of biological therapies, including growth factor antagonists and peptide mimetics, are also discussed.
Collapse
Affiliation(s)
- Daniel Thomas
- The Hanson Institute, Division of Human Immunology, The Institute of Medical and Veterinary Science, Adelaide, SA, Australia
| | | | | |
Collapse
|
52
|
Neumann TK, Foote M. The development of supportive-care agents for patients with cancer. ACTA ACUST UNITED AC 2003; 9:397-417. [PMID: 14650936 DOI: 10.1016/s1387-2656(03)09011-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
As the population ages, a dramatic increase in the number of cases of cancer is expected and the need for supportive-care agents, those used to ameliorate some of the side effects of cancer or its treatment, becomes more urgent. At present, supportive-care products are available and new agents are being developed with novel mechanisms of action or modifications of existing agents that improve performance. Because of the urgent need for such products, efficient development is required to deliver useful products to patients as rapidly as possible. This chapter uses actual examples to illustrate the stages of drug development, phase I through phase 3.
Collapse
Affiliation(s)
- Theresa K Neumann
- Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1699, USA
| | | |
Collapse
|
53
|
Farese AM, MacVittie TJ, Roskos L, Stead RB. Hematopoietic recovery following autologous bone marrow transplantation in a nonhuman primate: effect of variation in treatment schedule with PEG-rHuMGDF. Stem Cells 2003; 21:79-89. [PMID: 12529554 DOI: 10.1634/stemcells.21-1-79] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mathematical modeling of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) pharmacokinetics (PK) and pharmacodynamics (PD) suggest that variations in the PEG-rHuMGDF treatment schedule could reduce the severity and duration of thrombocytopenia following myeloablation and bone marrow transplant (BMT). We tested this hypothesis in a rhesus monkey model of autologous (Au) bone marrow-derived mononuclear cell (BM-MNC) transplantation following lethal myeloablation. On day 0, animals were myeloablated by total body exposure to 920 cGy, 250 kVp x-irradiation (TBI). Four cohorts of animals were infused with 1 x 10(8) AuBM-MNC/kg body weight within 2 hours of TBI. The AuBMT-alone cohort received no cytokine, the daily dosage cohort received PEG-rHuMGDF (2.5 micro g/kg/day, s.c.) post TBI and AuBMT, and the pre/post-transplant cohort received PEG-rHuMGDF (2.5 micro g/kg/day, s.c.) pre (day -9 to day -5) and post TBI and AuBMT. The post-transplant PEG-rHuMGDF administration in the above cohorts was begun on day 1 post TBI and continued until platelet counts reached 200,000 micro l (range, 15-31 days). Another group received PEG-rHuMGDF (300 micro g/kg/day, s.c.) on days 1 and 3 only following TBI and AuBMT. The TBI controls received neither AuBMT nor cytokine therapy. In this model of AuBMT, with regard to the PEG-rHuMGDF administration schedule, the daily dosage of the post-transplant cohort did not significantly improve platelet recovery; the pre/post-transplant schedule and an abbreviated high-dosage, post-transplant schedule (days 1 and 3) significantly improved the duration and nadir of thrombocytopenia and platelet recovery. These data confirm predictions from PK/PD modeling of PEG-rHuMGDF that thrombocytopenia is preventable following AuBMT.
Collapse
Affiliation(s)
- Ann M Farese
- University of Maryland Greenebaum Cancer Center, Baltimore, Maryland 21201, USA.
| | | | | | | |
Collapse
|
54
|
Kizaki M, Miyakawa Y, Ikeda Y. Long-term administration of pegylated recombinant human megakaryocyte growth and development factor dramatically improved cytopenias in a patient with myelodysplastic syndrome. Br J Haematol 2003; 122:764-7. [PMID: 12930386 DOI: 10.1046/j.1365-2141.2003.04504.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To date, there are no curative therapeutic options for patients with myelodysplastic syndromes (MDS) other than allogeneic stem cell transplantation. We treated an MDS patient with 10 microg/kg pegylated recombinant human megakaryocyte growth and development factor (rHuMGDF) for more than 450 d. The patient's platelet counts increased from <10 x 10(9)/l to 50 x 10(9)/l. Interestingly, haemoglobin levels increased dramatically and reached over 13 g/dl without additional transfusion. Adverse events and neutralizing antibodies were not observed during treatment, suggesting that the long-term administration of rHuMGDF might be of clinical benefit to patients with MDS.
Collapse
Affiliation(s)
- Masahiro Kizaki
- Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | | | | |
Collapse
|
55
|
Phillips KA, Bernhard J. Adjuvant breast cancer treatment and cognitive function: current knowledge and research directions. J Natl Cancer Inst 2003; 95:190-7. [PMID: 12569140 DOI: 10.1093/jnci/95.3.190] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Evidence is mounting that potentially curative systemic adjuvant therapy for early-stage breast cancer may result in cognitive impairment. Five published studies have investigated cognitive function in this setting, and the consistent results of all five studies suggest an adverse effect of adjuvant chemotherapy. These studies are reviewed with particular attention to their methodologic limitations. For example, all five studies used cross-sectional designs, none controlled for possible confounding hormonal factors, and three examined patients who had not received a uniform chemotherapy regimen. The potential roles of chemotherapy-induced menopause and of adjuvant hormonal therapy in cognitive impairment are also discussed. Priorities for future research include confirmation of an effect of adjuvant chemotherapy in a study with a longitudinal design, closer examination of the potential contribution of hormonal factors, and similar studies on the effect of adjuvant therapy on cognitive function in other cancer types. If an effect of systemic adjuvant therapy on cognitive function is confirmed, such an effect will have implications for informed consent. It may also result in incorporation of objective measures of cognition in clinical trials of adjuvant therapy and in the investigation of preventive interventions that might minimize the impact of cognitive dysfunction after cancer treatment.
Collapse
Affiliation(s)
- Kelly-Anne Phillips
- Department of Haematology and Medical Oncology, Peter MacCallum Cancer Institute, St. Andrew's Place, East Melbourne, Victoria, Australia.
| | | |
Collapse
|
56
|
Kuter DJ, Begley CG. Recombinant human thrombopoietin: basic biology and evaluation of clinical studies. Blood 2002; 100:3457-69. [PMID: 12411315 DOI: 10.1182/blood.v100.10.3457] [Citation(s) in RCA: 256] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Thrombocytopenia is a common medical problem for which the main treatment is platelet transfusion. Given the increasing use of platelets and the declining donor population, identification of a safe and effective platelet growth factor could improve the management of thrombocytopenia. Thrombopoietin (TPO), the c-Mpl ligand, is the primary physiologic regulator of megakaryocyte and platelet development. Since the purification of TPO in 1994, 2 recombinant forms of the c-Mpl ligand--recombinant human thrombopoietin (rhTPO) and pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF)--have undergone extensive clinical investigation. Both have been shown to be potent stimulators of megakaryocyte growth and platelet production and are biologically active in reducing the thrombocytopenia of nonmyeloablative chemotherapy. However, neither TPO has demonstrated benefit in stem cell transplantation or leukemia chemotherapy. Other clinical studies have investigated the use of TPO in treating chronic nonchemotherapy-induced thrombocytopenia associated with myelodysplastic syndromes, idiopathic thrombocytopenic purpura, thrombocytopenia due to human immunodeficiency virus, and liver disease. Based solely on animal studies, TPO may be effective in reducing surgical thrombocytopenia and bleeding, ex vivo expansion of pluripotent stem cells, and as a radioprotectant. Ongoing and future studies will help define the clinical role of recombinant TPO and TPO mimetics in the treatment of chemotherapy- and nonchemotherapy-induced thrombocytopenia.
Collapse
Affiliation(s)
- David J Kuter
- Hematology/Oncology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | | |
Collapse
|
57
|
Abstract
A variety of agents are available to improve hemostasis and reduce blood loss in multiple clinical settings. These agents are most commonly used to reduce bleeding when an underlying hemostatic defect is present. Some new agents offer the potential to decrease blood loss even in the absence of an obvious underlying hemostatic defect. The authors discuss the use of a variety of products to reduce bleeding and minimize transfusion of blood products in the setting of clotting factor deficiency or inhibition, platelet deficiency and/or dysfunction, increased fibrinolysis, therapeutic anticoagulation, and coagulopathies caused by dilution and consumption in the setting of trauma and surgery. The authors primarily focus on the available pharmaceuticals.
Collapse
|
58
|
Shibuya K, Kuwaki T, Tahara E, Yuki C, Akahori H, Kato T, Miyazaki H. Marked improvement of thrombocytopenia in a murine model of idiopathic thrombocytopenic purpura by pegylated recombinant human megakaryocyte growth and development factor. Exp Hematol 2002; 30:1185-92. [PMID: 12384150 DOI: 10.1016/s0301-472x(02)00898-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We examined the stimulatory effect of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) on platelet production in male (NZW x BXSB) F(l) (W/B F(1)) mice, a murine model of idiopathic thrombocytopenic purpura. MATERIALS AND METHODS A cohort of 19- to 25-week-old, severely thrombocytopenic male W/B F(1) mice were given PEG-rHuMGDF at different dosing schedules. Before and at various times after therapy, platelet counts, reticulated platelets, platelet lifespan, and levels of platelet-associated immunoglobulin G were measured. Analysis of megakaryocytic cells was performed. RESULTS Treatment of male W/B F(1) mice with PEG-rHuMGDF (30 microg/kg/day) three times per week for several weeks resulted in sustained thrombocytosis, accompanied by increased megakaryocytopoiesis in both the bone marrow and spleen. The degree of the platelet response to PEG-rHuMGDF varied between individual mice, likely reflecting the heterogeneity of the disease. Production of new platelets in response to PEG-rHuMGDF was manifested by an increase in reticulated platelets. Levels of platelet-associated immunoglobulin G decreased inversely during periods of thrombocytosis. PEG-rHuMGDF therapy also improved thrombocytopenia in male W/B F(1) mice refractory to splenectomy. Platelet lifespan was not affected by PEG-rHuMGDF. Male W/B F(1) mice treated with pegylated murine MGDF, a homologue of PEG-rHuMGDF, had persistent thrombocytosis for at least 7 months, suggesting that antiplatelet antibody production was not enhanced. CONCLUSIONS PEG-rHuMGDF therapy potently stimulated platelet production, effectively ameliorating thrombocytopenia in a murine model of idiopathic thrombocytopenic purpura.
Collapse
Affiliation(s)
- Kazunori Shibuya
- Pharmaceutical Development Laboratory, Kirin Brewery Company, Ltd., Gunma, Japan
| | | | | | | | | | | | | |
Collapse
|
59
|
Schuster MW, Beveridge R, Frei-Lahr D, Abboud CN, Cruickshank S, Macri M, Menchaca D, Holden J, Waller EK. The effects of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) on platelet recovery in breast cancer patients undergoing autologous bone marrow transplantation. Exp Hematol 2002; 30:1044-50. [PMID: 12225796 DOI: 10.1016/s0301-472x(02)00878-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To assess the safety and efficacy of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) administered after autologous bone marrow transplantation (ABMT). PATIENTS AND METHODS Two randomized, double-blind, placebo-controlled studies were done. In the phase 1/2 study, 75 breast cancer patients underwent a bone marrow harvest and myeloablative STAMP V chemotherapy and were randomized to receive placebo or one of three doses of PEG-rHuMGDF. In the phase 3 study, 64 patients were randomized to receive placebo or the minimally effective dose of PEG-rHuMGDF. The study drug was administered daily starting on the day of bone marrow infusion until the platelet count was greater than or equal to 50 x 10(9)/L (without transfusion) or for a maximum of 28 days. All patients received 10 microg/kg/day filgrastim starting on day 2 until neutrophil count recovery. RESULTS PEG-rHuMGDF appeared to be safe and well tolerated. No significant differences were noted in mortality or disease progression rates. Antibodies to MGDF were not observed. In the phase 1/2 study, the time to platelet recovery to greater than or equal to 20 x 10(9)/L and platelet transfusion requirements were significantly reduced for patients treated with PEG-rHuMGDF compared with placebo (p < 0.05). In the phase 3 study, no significant differences in the kinetics of early thrombopoiesis or platelet transfusions after ABMT were observed. CONCLUSIONS PEG-rHuMGDF was not consistently efficacious in reducing the duration of severe thrombocytopenia. The maximum platelet counts for PEG-rHuMGDF-treated patients occurred a median of 2 weeks after the last dose of drug, suggesting that the biologic effects of this hematopoietic cytokine are delayed compared with other hematopoietic cytokines.
Collapse
|
60
|
Haznedaroglu IC, Goker H, Turgut M, Buyukasik Y, Benekli M. Thrombopoietin as a drug: biologic expectations, clinical realities, and future directions. Clin Appl Thromb Hemost 2002; 8:193-212. [PMID: 12361196 DOI: 10.1177/107602960200800301] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
After the cloning of thrombopoietin (c-mpl ligand, Tpo) in 1994, 2 recombinant thrombopoietic growth factors, full-length glycosylated recombinant human Tpo (reHuTPO) and polyethylene glycol conjugated megakaryocyte growth and development factor (PEG-reHuMGDF), have been studied in humans in a variety of clinical settings. Both thrombopoietins are generally well tolerated if administered intravenously (IV). The c-mpl ligands produce a dose-related enhancement of platelet levels, reduce nonmyeloablative chemotherapy-induced mild thrombocytopenia, and mobilize hematopoietic progenitors. On September 11, 1998, the development of PEG-reHuMGDF was suspended in the U.S., due to formation of the neutralizing anti-Tpo antibody. Those neutralizing antibodies lead to thrombocytopenia and pancytopenia in some patients receiving subcutaneous (SC) PEG-reHuMGDF. Japanese investigators indicate that the probability of antibody formation against PEG-reHuMGDF is low when the drug is administered IV instead of SC. reHuTPO has a more favorable safety profile from the point of antibody production. The c-mpl ligands can improve apheresis yields when administered to normal platelet donors. Preliminary data about the use of PEG-reHuMGDF in myelodysplasia, aplastic anemia, and immune thrombocytopenic purpura are promising. Tpo is usually not effective in myeloablative thrombocytopenia when bone marrow hematopoietic progenitors are not present. The major obstacle for the thrombopoietins is their delayed action for managing clinical thrombocytopenia. This review will focus on the biologic basis, current clinical experience, and future directions for the use of thrombopoietic molecules as drugs. The identification of a safe, effective, and potent pharmacologic platelet growth factor could significantly improve the management of thrombocytopenia-induced bleeding.
Collapse
|
61
|
Partridge AH, Burstein HJ, Winer EP. Side effects of chemotherapy and combined chemohormonal therapy in women with early-stage breast cancer. J Natl Cancer Inst Monogr 2002:135-42. [PMID: 11773307 DOI: 10.1093/oxfordjournals.jncimonographs.a003451] [Citation(s) in RCA: 246] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The decision to receive chemotherapy or chemohormonal therapy involves careful consideration of both the potential benefits and possible risks of therapy. There are substantial short- and long-term side effects from chemotherapy. By convention, short-term side effects include those toxic effects encountered during chemotherapy, while long-term side effects include later complications of treatment arising after the conclusion of adjuvant chemotherapy. These side effects vary, depending on the specific agents used in the adjuvant regimen as well as on the dose used and the duration of treatment. There is also considerable variability in side effect profile across individuals. This review will focus on the short- and long-term toxicity seen with the most commonly used adjuvant chemotherapy and chemohormonal therapy regimens.
Collapse
Affiliation(s)
- A H Partridge
- Breast Oncology Center, Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02215, USA
| | | | | |
Collapse
|
62
|
Simes RJ, Coates AS. Patient preferences for adjuvant chemotherapy of early breast cancer: how much benefit is needed? J Natl Cancer Inst Monogr 2002:146-52. [PMID: 11773309 DOI: 10.1093/oxfordjournals.jncimonographs.a003453] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Adjuvant chemotherapy for early-stage breast cancer has been shown to delay recurrence and improve survival. However, the benefits are modest and must be balanced against the adverse treatment effects. We assessed the size of the survival benefit needed to justify the toxicity of chemotherapy, based on the preferences of women who had previously received adjuvant cyclophosphamide, methotrexate, and 5-fluorouracil (CMF). We also attempted to identify circumstances in which larger survival gains would be needed. In semistructured interviews, 104 women who had received adjuvant CMF chemotherapy were asked to rate the survival benefit that would justify 6 months of such treatment, using a series of hypothetical trade-offs between shorter survival without treatment and longer survival with treatment. Similar preferences were sought for a greater probability of 5-year survival. Most patients considered 6 months of adjuvant CMF chemotherapy worthwhile for relatively modest survival gains: 77% considered an increase of from 5 to 6 years worthwhile, 74% thought an increase of from 15 to 17 years worthwhile, and more than 70% considered such treatment justified for a 5% greater chance of living 5 or more years. Smaller survival benefits were needed for women who had experienced less toxicity (P =.01), had not received initial radiotherapy (P =.01), had better social support (P =.02), and had others at home dependent on their support (P =.0001). Modest survival benefits are sufficient to justify adjuvant cytotoxic chemotherapy for most women with early-stage breast cancer. Individual preferences are important when weighing trade-offs between survival and adverse treatment effects.
Collapse
Affiliation(s)
- R J Simes
- R. J. Simes, National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, New South Wales 2006, Australia.
| | | |
Collapse
|
63
|
Abstract
Megakaryopoiesis and subsequent thrombopoiesis occur through complex biologic steps: megakaryocyte precursors that developed from hematopoietic stem cells initially proliferate, then differentiate into mature polyploid megakaryocytes, and finally release platelets. Although a number of growth factors can augment megakaryopoiesis in vitro, thrombopoietin is a physiologic and the most potent regulator of megakaryopoiesis in vitro and in vivo. Thrombopoietin induces the growth of megakaryocyte precursors through activation of multiple signaling cascades, including Ras/mitogen-activated protein kinase (MAPK), signal transducers and activators of transcription 5 (STAT5), phosphatidylinositol 3-kinase (PI3-K)/Akt, and protein kinase C, whereas it induces megakaryocytic maturation primarily through the Ras/MAPK pathway. During the maturation step, megakaryocytes undergo polyploidization characterized by repeated rounds of DNA replication without concomitant cell division. During these rounds of replication, cytokinesis is neglected because of the down-regulated expression of AIM-1, and DNA replication occurs through the increased expression of D-type cyclins. As for transcriptional regulation during megakaryopoiesis, GATA-1 plays a central role in the lineage commitment of hematopoietic stem cells toward erythroid/megakaryocytic lineage and subsequent maturation. p45 NF-E2 is essential for platelet release from terminally differentiated megakaryocytes. At present, mutations of GATA-1, AML1, and HOXA11 genes have been found in hereditary diseases accompanying thrombocytopenia among humans.
Collapse
Affiliation(s)
- Itaru Matsumura
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | | |
Collapse
|
64
|
Abstract
Cytokines are a growing group of proteins that are responsible for the communication of cells of the immune system, hematopoietic cells, and other cell types. They play a dominant role in various diseases, particularly in promoting and perpetuating inflammation. Cytokine production is a reaction of the body to a pathologic state to restore homeostasis. In such cases, the therapeutic intervention should support the reaction of the body by giving the cytokine itself (agonistic therapeutics). In other cases, manifestation of a disease results from an overproduction of cytokines, making cytokine antagonists desirable therapeutic drugs. Furthermore, cytokines may be good candidates as cancer therapeutics, especially to support the restoration of blood cell populations after chemotherapy or radiation.
Collapse
Affiliation(s)
- Heidi Schooltink
- Department of Biochemistry, Christian-Albrechts-Universität Kiel, D-24098 Kiel, Germany
| | | |
Collapse
|
65
|
Basser RL, O'Flaherty E, Green M, Edmonds M, Nichol J, Menchaca DM, Cohen B, Begley CG. Development of pancytopenia with neutralizing antibodies to thrombopoietin after multicycle chemotherapy supported by megakaryocyte growth and development factor. Blood 2002; 99:2599-602. [PMID: 11895799 DOI: 10.1182/blood.v99.7.2599] [Citation(s) in RCA: 199] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Clinical trials of thrombopoietin (TPO), the central regulator of megakaryocytopoiesis, have revealed few side effects associated with its use. We here report a case of pancytopenia associated with the development of neutralizing antibodies to TPO that occurred in a patient who had undergone multicycle chemotherapy with multiple cycles of subcutaneous administration of pegylated recombinant human megakaryocyte growth and development factor. Samples of the patient's bone marrow showed trilineage hypoplasia with absence of myeloid, erythroid, and megakaryocyte progenitor cells but with elevated endogenous levels of erythropoietin, granulocyte colony-stimulating factor, and stem-cell factor. To our knowledge, this is the first report of an aplastic anemia-like syndrome associated with neutralizing antibodies to TPO.
Collapse
Affiliation(s)
- Russell L Basser
- Centre for Developmental Cancer Therapeutics, Department of Haematology and Medical Oncology, Ludwig Institute for Cancer Research, Royal Melbourne Hospital, 45 Poplar Road, Parkville, Victoria 3052, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
66
|
|
67
|
Abstract
The recent discovery of thrombopoietin has enhanced our understanding of both hematopoiesis and platelet production. Thrombopoietin supports hematopoietic stem cell survival and expansion as well as promoting all aspects of megakaryocyte development. The hormone displays many structural similarities to other members of the hematopoietic cytokine family and some notable differences, and regulation of its expression requires both receptor-mediated removal and other mechanisms. Thrombopoietin induces receptor dimerization and tyrosine phosphorylation, and a series of signaling events including activation of JAK/STAT, Shc/Ras/MAPK and PI3K/Akt; these pathways overlap with those induced by other cytokines, but the differences that lead to the unique biological effects of the hormone are gradually being uncovered. Our growing appreciation of how cytokine signaling pathways are translated into megakaryocyte development is discussed.
Collapse
Affiliation(s)
- Amy E Geddis
- Division of Hematology, University of Washington School of Medicine, Box 357710, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | | | | |
Collapse
|
68
|
Matsubara K, Baba K, Nigami H, Harigaya H, Ishiguro A, Kato T, Miyazaki H. Early elevation of serum thrombopoietin levels and subsequent thrombocytosis in healthy preterm infants. Br J Haematol 2001; 115:963-8. [PMID: 11843834 DOI: 10.1046/j.1365-2141.2001.03183.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
To verify pathophysiological mechanisms underlying thrombocytosis in low-birth-weight (LBW) preterm babies, we evaluated kinetic changes in platelet counts and thrombopoietic cytokines including thrombopoietin (TPO), interleukin 6 (IL-6) and IL-11 in 24 uncomplicated preterm infants. Platelet counts in cord blood (CB) (265 +/- 64 x 10(9)/l) were similar to adult levels, increased by d 14 (473 +/- 140 x 10(9)/l), and then remained fairly constant. Thrombocytosis (> 500 x 10(9)/l) was observed in 9/24 (38%) subjects. Mean TPO level in CB was 5.11 +/- 1.51 fmol/ml, peaked at d 2 (7.64 +/- 3.28 fmol/ml), decreased at d 5 (3.93 +/- 1.67 fmol/ml), and thereafter kept fairly constant during the remaining neonatal period. Compared with term infants, mean TPO levels of preterm infants in CB and at d 2 were significantly higher (P < 0.01). There was an inverse correlation between platelet counts and TPO levels (r = 0.45, P < 0.001, n = 88). Preterm neonates with thrombocytosis had significantly higher TPO values in CB than those without thrombocytosis (P < 0.05). There was no significant relationship between platelet counts and IL-6. IL-11 was not detectable. These results suggest that an early elevation of serum TPO levels is related to the subsequent thrombocytosis in LBW preterm infants.
Collapse
Affiliation(s)
- K Matsubara
- Department of Paediatrics, Nishi-Kobe Medical Centre, 5-7-1 Kojidai, Nishi-ku, Kobe 651-2273, Japan.
| | | | | | | | | | | | | |
Collapse
|
69
|
Basser RL, Begley CG. Failing to live up to the fanfare? A personal perspective on obstacles to the clinical development of thrombopoietic agents. Int J Hematol 2001; 74:390-6. [PMID: 11794693 DOI: 10.1007/bf02982081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A number of hematopoietic growth factors have been identified that are active on megakaryocytes and platelets, but only 2, interleukin-11 (IL-11) and thrombopoietin, are being actively pursued clinically, with IL-11 approved for treatment of thrombocytopenia. The development of these agents in general has been disappointing, and in part this reflects the inherent biology of these factors with a failure to match clinical need with physiological function. The delayed action of these factors is also a consequence of the intrinsic biology of megakaryocytes and platelets, and thus is likely to be limiting regardless of which factor is employed. In addition, the development of these agents has occurred at a time when there is something of a decreasing demand for platelets, at least in the context of chemotherapy-induced thrombocytopenia. This decrease is the result of increased use of blood stem cells to support intensive chemotherapy procedures, reduced thresholds for platelet transfusion, and a decreasing role for intensive chemotherapy. These issues are discussed.
Collapse
Affiliation(s)
- R L Basser
- The Centre for Developmental Cancer Therapeutics, Parkville, Victoria, Australia
| | | |
Collapse
|
70
|
Kaser A, Brandacher G, Steurer W, Kaser S, Offner FA, Zoller H, Theurl I, Widder W, Molnar C, Ludwiczek O, Atkins MB, Mier JW, Tilg H. Interleukin-6 stimulates thrombopoiesis through thrombopoietin: role in inflammatory thrombocytosis. Blood 2001; 98:2720-5. [PMID: 11675343 DOI: 10.1182/blood.v98.9.2720] [Citation(s) in RCA: 449] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Baseline platelet production is dependent on thrombopoietin (TPO). TPO is constitutively produced and primarily regulated by receptor-mediated uptake by platelets. Inflammatory thrombocytosis is thought to be related to increased interleukin-6 (IL-6) levels. To address whether IL-6 might act through TPO to increase platelet counts, TPO was neutralized in vivo in C57BL/10 mice treated with IL-6, and hepatic TPO mRNA expression and TPO plasma levels were studied. Transcriptional regulation of TPO mRNA was studied in the hepatoblastoma cell line HepG2. Furthermore, TPO plasma levels were determined in IL-6-treated cancer patients. It is shown that IL-6-induced thrombocytosis in C57BL/10 mice is accompanied by enhanced hepatic TPO mRNA expression and elevated TPO plasma levels. Administration of IL-6 to cancer patients results in a corresponding increase in TPO plasma levels. IL-6 enhances TPO mRNA transcription in HepG2 cells. IL-6-induced thrombocytosis can be abrogated by neutralization of TPO, suggesting that IL-6 induces thrombocytosis through TPO. A novel pathway of TPO regulation by the inflammatory mediator IL-6 is proposed, indicating that the number of platelets by themselves might not be the sole determinant of circulating TPO levels and thus of thrombopoiesis. This regulatory pathway might be of relevance for the understanding of reactive thrombocytosis.
Collapse
Affiliation(s)
- A Kaser
- Divisions of Gastroenterology and Hepatology and of General Internal Medicine, the Department of Medicine, University Hospital Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Kuter DJ, Goodnough LT, Romo J, DiPersio J, Peterson R, Tomita D, Sheridan W, McCullough J. Thrombopoietin therapy increases platelet yields in healthy platelet donors. Blood 2001; 98:1339-45. [PMID: 11520780 DOI: 10.1182/blood.v98.5.1339] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The recombinant thrombopoietins have been shown to be effective stimulators of platelet production in cancer patients. It was therefore of interest to determine if one of these, pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF), could be used to increase platelet counts and consequently platelet yields from apheresis in healthy platelet donors. In a blinded, 2-cycle, crossover study, 59 platelet donors were randomized to receive a single subcutaneous injection of PEG-rHuMGDF (1 microg/kg or 3 microg/kg) or placebo and 15 days later undergo platelet apheresis. Donors treated with placebo had a median peak platelet count after PEG-rHuMGDF injection of 248 x 10(9)/L compared with 366 x 10(9)/L in donors treated with 1 microg/kg PEG-rHuMGDF and 602 x 10(9)/L in donors treated with 3 microg/kg PEG-rHuMGDF. The median maximum percentage that platelet counts increased from baseline was 10% in donors who received placebo compared with 70% in donors who received 1 microg/kg and 167% in donors who received 3 microg/kg PEG-rHuMGDF. There was a direct relationship between the platelet yield and the preapheresis platelet count: Placebo-treated donors provided 3.8 x 10(11) (range 1.3 x 10(11)-7.9 x 10(11)) platelets compared with 5.6 x 10(11) (range 2.6 x 10(11)-12.5 x 10(11)) or 11.0 x 10(11) (range 7.1 x 10(11)-18.3 x 10(11)) in donors treated with 1 microg/kg or 3 microg/kg PEG-rHuMGDF, respectively. Substandard collections (<3 x 10(11) platelets) were obtained from 26%, 4%, and 0% of the placebo, 1 microg/kg, and 3 microg/kg donors, respectively. No serious adverse events were reported; nor were there events that met the criteria for dose-limiting toxicity. Thrombopoietin therapy can increase platelet counts in healthy donors to provide a median 3-fold more apheresis platelets compared with untreated donors.
Collapse
Affiliation(s)
- D J Kuter
- Hematology/Oncology Unit, Massachusetts General Hospital, Boston 02114, USA.
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Ishii T, Futami S, Nishida M, Suzuki T, Sakamoto T, Suzuki N, Maekawa K. Brief note and evaluation of acute-radiation syndrome and treatment of a Tokai-mura criticality accident patient. JOURNAL OF RADIATION RESEARCH 2001; 42 Suppl:S167-S182. [PMID: 11791750 DOI: 10.1269/jrr.42.s167] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Patient A who was exposed to a critical dose of radiation developed skin lesions throughout the body surface, gastrointestinal disorder with massive diarrhea and prominent bleeding, which caused severe loss in body fluids. Gastrointestinal bleeding due to the deteriorated intestinal mucosa was considered to be one of the major causes of death, although infection did not develop, possibly because of SDD and aseptic intensive care, until terminal stages. Patient A ultimately developed respiratory and renal failure in addition to skin exudate and gastrointestinal bleeding, and died of multiple organ failure on the 83rd day after exposure. The extreme unevenness of the dose distribution and the neutron versus y-ray component made the clinical manifestation very complicated. Initially, the mean absorbed dose was calculated as 16-20 GyEq for Patient A, mainly based on neutron-activated 24Na in the blood. However, a very recent calculation showed that the absorbed skin dose was highest at the upper-right abdomen reaching 61.8 Gy (27.0 as neutron plus 34.8 Gy as y-ray). The dorsal side was calculated to have received one eighth of the value of the abdominal side, and much smaller neutron component. His absorbed-dose distribution throughout the body was very inhomogeneous because of the closeness of the standing point to the mixing tank. Despite prolonged survival because of intensive care with massive fluids and blood transfusion, peripheral blood stem-cell transplantation, cultured skin-cell grafts, and the administration of cytokines for marrow, the patient was not saved. Restoration of the bone marrow function, prevention of skin fibrosis, radiation lung damage, and repair of gastrointestinal mucosa, and final recovery of the patient were elusive. Abundant personnel and resources were also a prerequisite to allow for the comprehensive and collective intensive care. A further understanding of the effects of high-dose radiation as well as the basic and clinical development of regeneration medicine are important issues for the future.
Collapse
Affiliation(s)
- T Ishii
- The Department of Traumatology and Critical Care, Graduate School of Medicine, University of Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
73
|
Affiliation(s)
- R L Basser
- Centre for Developmental Cancer Therapeutics, Parkville, Victoria, Australia.
| | | |
Collapse
|
74
|
Honda K, Takenaka K, Shinagawa K, Ishimaru F, Ikeda K, Niiya K, Harada M. Synergistic effects of pegylated recombinant human megakaryocyte growth and development factor and granulocyte colony-stimulating factor on mobilization of hematopoietic progenitor and stem cells with long-term repopulating ability into peripheral blood in mice. Bone Marrow Transplant 2001; 28:329-34. [PMID: 11571503 DOI: 10.1038/sj.bmt.1703140] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2001] [Accepted: 06/05/2001] [Indexed: 11/09/2022]
Abstract
We investigated the effects of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) on peripheral blood progenitor cell (PBPC) mobilization and the combined effect of PEG-rHuMGDF plus recombinant human granulocyte colony-stimulating factor (rhG-CSF) in C57BL/6 mice. Treatment of mice with PEG-rHuMGDF increased the numbers of day 8 and day 12 spleen colony-forming units (CFU-S), and pre-CFU-S in the PB. Ten days administration of PEG-rHuMGDF could mobilize higher numbers of days 8 and day 12 CFU-S than 5 days administration. An optimal dose of PEG-rHuMGDF mobilized a higher number of committed progenitor cells (day 8 CFU-S) and a lower number of immature progenitor cells (pre-CFU-S) into PB than rhG-CSF. The combined administration of optimal or suboptimal doses of PEG-rHuMGDF and rhG-CSF induced synergistic effects on mobilization of CFU-S and pre-CFU-S into PB compared to either factor alone. Four months after sex-mismatched PBPC transplantation, long-term donor-derived engraftment was observed in all recipients that had been transplanted with PBPCs mobilized by rhG-CSF and/or PEG-rHuMGDF, as determined by Y-chromosome polymerase chain reaction (PCR) analysis. Our data suggest that cytokine-induced pathways for PBPC mobilization may be different between PEG-rHuMGDF and rhG-CSF and indicate that PEG-rHuMGDF alone or the combination with rhG-CSF may be useful in effective PBPC mobilization.
Collapse
Affiliation(s)
- K Honda
- Department of Internal Medicine II, Okayama University Medical School, Okayama, Japan
| | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
The relative merits of apheresis platelets and platelet concentrates are undergoing debate due to evolving issues of safety, inventory, and cost. The application of photochemical inactivation technology may eliminate any rationale for the use of apheresis platelets rather than pooled platelet concentrates, so that the relative merits of these two alternatives will be debated over costs and inventory. Doses of apheresis platelets are determined by donor platelet count and by platelet yield. The generation of a platelet apheresis inventory has been accompanied by a decline in whole blood inventory; research into the impact of these distinct donor pools on national blood policy is needed.
Collapse
Affiliation(s)
- L T Goodnough
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| |
Collapse
|
76
|
Abstract
Recombinant haemopoietic growth factors (HGFs) are an attractive adjunct to reduce morbidity from chemotherapy regimens and their use has become widespread in paediatric oncology. Although patients receiving HGFs often have faster haematological recovery after intensive chemotherapy, this does not always translate into meaningful clinical benefits. This article reviews the clinical effectiveness of HGFs in a variety of different contexts. Most published studies have used granulocyte colony-stimulating factor (G-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF) as prophylaxis to ameliorate the subsequent neutropenia following intensive chemotherapy. These 2 agents have also been used to mobilise peripheral blood stem cells for autologous transplantation. HGFs specific for anaemia and thrombocytopenia are currently in paediatric clinical trials and it is hoped that the proper context and administration strategy can be found to make their use clinically effective. This article also reviews data on toxicity, specifically focusing on differences between various formulations of growth factors. HGFs are expensive, and cost-benefit analyses reviewed in this article give an important perspective on the financial aspects of paediatric cancer care. Because HGFs do not benefit every child receiving chemotherapy and overuse increases costs and may result in unnecessary adverse effects, evidence-based guidelines for their rational use in paediatric oncology are proposed.
Collapse
Affiliation(s)
- L M Wagner
- Department of Hematology/Oncology, St Jude Children's Research Hospital, Memphis, Tennessee 38105-2794, USA
| | | |
Collapse
|
77
|
Abstract
The liver plays an important role in the production of haemopoietic hormones. It acts as the primary site of synthesis of erythropoietin (EPO) in the fetal stage, and it is the predominant thrombopoietin (TPO)-producing organ for life. In contrast to that of EPO and other liver proteins, the hepatic synthesis of TPO is influenced little by external signals. Hepatocytes express the TPO gene in a constitutive way, i.e. irrespective of the level of platelets in blood. Megakaryocytes and platelets remove the hormone from blood by means of their high-affinity TPO receptors. Normally, the plasma level of TPO is relatively low ( approximately 10(-12) mol/l). However, in thrombocytopenic states due to marrow failure or bleeding, the concentration of circulating TPO may increase greatly. The simple feedback regulation by TPO and its target cells is efficient in maintaining constant platelet numbers in healthy people. Persisting thrombocytopenia develops only in severe liver or marrow failure. On the other hand, an increase in circulating TPO and interleukin 6 (IL-6) may cause reactive thrombocytosis in inflammatory diseases, including cancer. The indications for recombinant human thrombopoietin (rHuTPO) therapy and its impact on transfusion medicine are still under investigation.
Collapse
Affiliation(s)
- W Jelkmann
- Institute of Physiology, Medical University of Lubeck, Lubeck, Germany.
| |
Collapse
|
78
|
Del Vecchio A, Sola MC, Theriaque DW, Hutson AD, Kao KJ, Wright D, Garcia MG, Pollock BH, Christensen RD. Platelet transfusions in the neonatal intensive care unit:factors predicting which patients will require multiple transfusions. Transfusion 2001; 41:803-8. [PMID: 11399824 DOI: 10.1046/j.1537-2995.2001.41060803.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Previous studies suggest that recombinant thrombopoietin (rTPO) will increase platelet production in thrombocytopenic neonates. However, the target populations of neonates most likely to benefit should be defined. Studies suggest that rTPO will not elevate the platelet count until 5 days after the start of treatment. Therefore, the neonates who might benefit from rTPO are those who will require multiple platelet transfusions for more than 5 days. This study was designed to find means of prospectively identifying these patients. STUDY DESIGN AND METHODS A historic cohort study of all patients in the neonatal intensive care unit (NICU) at the University of Florida who received platelet transfusions from January 1, 1997, through December 31, 1998, was conducted. RESULTS Of the 1389 patients admitted to the NICU during the study period, 131 (9.4%) received platelet transfusions. Seventeen were treated with extracorporeal membrane oxygenation and were excluded from further analysis. Of the remaining 114 patients, 55 (48%) received one transfusion and 59 (52%) received more than one transfusion (21 had >4). None of the demographic factors examined predicted multiple platelet transfusions. However, two clinical conditions did; liver disease and renal insufficiency. Neonates who received one platelet transfusion had a relative risk of death 10.4 times that in neonates who received none (p = 0.0001). Neonates who received >4 platelet transfusions had a risk of death 29.9 times that in those who received no transfusions (p = 0.0001). CONCLUSION NICU patients with liver disease or renal insufficiency who receive one platelet transfusion are likely to receive additional transfusions. Therefore, these patients constitute a possible study population for a Phase I/II rTPO trial.
Collapse
Affiliation(s)
- A Del Vecchio
- Division of Neonatology, Department of Pediatrics, University of Florida, College of Medicine, Gainesville, Florida, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Sola MC, Dame C, Christensen RD. Toward a rational use of recombinant thrombopoietin in the neonatal intensive care unit. J Pediatr Hematol Oncol 2001; 23:179-84. [PMID: 11305723 DOI: 10.1097/00043426-200103000-00013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- M C Sola
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, USA.
| | | | | |
Collapse
|
80
|
Basser RL. Early Australian clinical studies with pegylated recombinant human megakaryocyte growth and development factor. Stem Cells 2001; 16 Suppl 2:225-9. [PMID: 11012194 DOI: 10.1002/stem.5530160725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF, an Mpl ligand) is a truncated form of native thrombopoietin currently undergoing clinical development. A series of studies in Australia have examined the safety and biological activities of PEG-rHuMGDF. Administration of PEG-rHuMGDF causes a dose-dependent increase in platelet count but has no effect on white cell count or hematocrit. These platelets are morphologically and functionally normal. When administered following moderately myelosuppressive chemotherapy, PEG-rHuMGDF significantly enhances platelet recovery, although scheduling in relation to chemotherapy may be important in optimizing the full effects. PEG-rHuMGDF mobilizes progenitor cells of multiple hematopoietic lineages, and alters the kinetics of peripheral blood progenitor cell mobilization after chemotherapy and filgrastim. PEG-rHuMGDF is well tolerated and does not cause toxicity similar to that observed with other thrombopoietic cytokines. Numerous studies are underway to help determine the precise role of PEG-rHuMGDF in clinical practice.
Collapse
Affiliation(s)
- R L Basser
- Centre for Development of Cancer Therapeutics, Parkville, Victoria, Australia
| |
Collapse
|
81
|
Taskin AL, Cohen-Solal K, Le Couedic JP, Abina MA, Villeval JL, Debili N, Casadevall N, Vainchenker W, Wendling F. Myelofibrosis: experimental models and human studies. Stem Cells 2001; 16 Suppl 2:155-64. [PMID: 11012187 DOI: 10.1002/stem.5530160718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Thrombopoietin (TPO) is the central regulator of megakaryocytopoiesis and thrombocytopoiesis. Preclinical data and human studies have so far shown that the recombinant molecule is safe to administer and associated with very little toxicity. Nevertheless, different experimental animal models have revealed that a chronic exposure to very high doses of TPO could result in myeloproliferative syndromes with a spectrum of pathological features in common with human idiopathic myelofibrosis (PMF). A number of investigators have researched whether TPO or its receptor Mpl were involved in the pathogenesis of human myeloproliferative syndromes which are also characterized by a predominant megakaryocytic involvement, in PMF and primitive essential thrombocythemia. In both diseases, megakaryocyte (MK) progenitors develop autonomously in serum-deprived cultures. This spontaneous MK development is also observed at limiting dilution demonstrating that MK escape the normal regulatory controls. Furthermore, this abnormal MK proliferation and maturation is neither due to an autocrine stimulation by TPO nor by point mutation or deletion in the coding region of the c-mpl gene. This paper will review the data that have been reported to date on the effects of an overexpression of Mpl ligand and related molecules on the induction of experimental myelofibrosis and highlight recent insights into the pathogenesis of PMF.
Collapse
Affiliation(s)
- A L Taskin
- Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Nichol JL. Endogenous TPO (eTPO) levels in health and disease: possible clues for therapeutic intervention. Stem Cells 2001; 16 Suppl 2:165-75. [PMID: 11012188 DOI: 10.1002/stem.5530160719] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The factor which is the primary regulator of megakaryocyte and platelet production has recently been identified as the ligand for the receptor Mpl. This discovery has resulted in substantial advances in our understanding of platelet homeostasis. The access to new experimental reagents has enabled studies of the endogenous circulating form of this ligand, endogenous thrombopoietin, in normal individuals and in patients with altered platelet numbers. The relationship of endogenous TPO in health and disease will be examined with consideration of the implications for successful therapeutic intervention with exogenous recombinant Mpl ligands in selected settings.
Collapse
Affiliation(s)
- J L Nichol
- Amgen, Inc., Thousand Oaks, California, USA
| |
Collapse
|
83
|
Sheridan W, Menchaca D. Overview of the safety and biologic effects of PEG-rHuMGDF in clinical trials. Stem Cells 2001; 16 Suppl 2:193-8. [PMID: 11012191 DOI: 10.1002/stem.5530160722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Completed randomized placebo-controlled phase I/II studies of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) have demonstrated that this recombinant Mpl ligand has potent and lineage-dominant effects on megakaryopoiesis and platelet production. Platelets produced after PEG-rHuMGDF administration display normal ultrastructure and functional attributes. In these early studies, PEG-rHuMGDF accelerated the recovery of baseline platelet counts after cytotoxic chemotherapy in cancer patients by six to seven days, indicating the potential for clinical benefit in this setting. PEG-rHuMGDF has been well-tolerated in clinical trials, with similar adverse events in placebo and PEG-rHuMGDF populations, and an observed adverse event profile consistent with the effects of underlying malignancy and chemotherapy. The lack of inflammatory cytokine effects in the clinic is consistent with results of animal studies, the narrow tissue distribution of Mpl and the lineage-dominant effect of PEG-rHuMGDF on megakaryopoiesis. Additional phase I/II studies have commenced in the fields of cancer chemotherapy and augmentation of platelet donation, and a phase III study is underway in patients undergoing bone marrow transplantation.
Collapse
Affiliation(s)
- W Sheridan
- Amgen Inc., Thousand Oaks, California, USA
| | | |
Collapse
|
84
|
Wolff SN, Herzig R, Lynch J, Ericson SG, Greer JP, Stein R, Goodman S, Benyunes MC, Ashby M, Jones DV, Fay J. Recombinant human thrombopoietin (rhTPO) after autologous bone marrow transplantation: a phase I pharmacokinetic and pharmacodynamic study. Bone Marrow Transplant 2001; 27:261-8. [PMID: 11277173 DOI: 10.1038/sj.bmt.1702772] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Thrombocytopenia following myelotoxic therapy is a common problem and when severe (<20,000/microl) can lead to severe morbidity and mortality. Thrombopoietin (TPO) is a naturally occurring glycosylated peptide which stimulates the differentiation of bone marrow stem cells into megakaryocyte progenitor cells, induces the expression of megakaryocyte differentiation markers, promotes megakaryocyte proliferation, polyploidization and, ultimately, the formation of increased numbers of platelets in the circulation. TPO has now been produced by recombinant technology and has entered clinical trials. This open label phase I study was designed to determine the safety, tolerance and pharmacokinetics of recombinant thrombopoietin (rhTPO) when administered to patients after undergoing high-dose chemotherapy followed by autologous bone marrow transplantation. rhTPO was administered intravenously by bolus injection at doses ranging from 0.3 to 4.8 microg/kg/day every 3 days to 30 patients and 0.6 microg/kg daily to three patients. rhTPO was begun the day after marrow infusion and continued until platelet recovery to >20,000/microl. G-CSF was concomitantly administered to promote myeloid recovery. Serious adverse events or neutralizing antibodies to rhTPO were not observed during the study. Median platelet recovery after ABMT was 19 days (range, 11-41). Neither the dose nor the schedule of rhTPO appeared to have any impact upon the time course of platelet recovery. In this phase I study, rhTPO was found to be well tolerated without the development of neutralizing antibodies and without compromising neutrophil recovery. Platelet recovery was similar for all doses studied warranting further evaluation in phase II and III trials designed to test for platelet recovery efficacy.
Collapse
Affiliation(s)
- S N Wolff
- Bone Marrow Transplant Program, Vanderbilt University, Nashville, TN 37232-5505, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Scott CL, Robb L, Nandurkar HH, Mansfield R, Alexander WS, Begley CG. Thrombopoietin signaling is required for in vivo expansion of IL-11--responsive hematopoietic progenitor cells in the steady state. Exp Hematol 2001; 29:138-45. [PMID: 11166452 DOI: 10.1016/s0301-472x(00)00622-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE mpl(-/-) mice have a profound defect in platelets and megakaryocytes and a defect in hematopoietic progenitor cells and stem cells. However, no specific subset of the progenitor/stem cell compartment has been shown to be particularly affected by this deficiency in mpl(-/-) mice. In this article, we identified a specific subset of bone marrow progenitor/stem cells that was altered in mpl(-/-) mice. MATERIALS AND METHODS In vitro and in vivo hematopoietic assays were utilized to examine the response to interleukin-11 in mice lacking the receptor for thrombopoietin (TPO) (mpl(-/-) mice). RESULTS The interleukin (IL)-11-responsive subset of progenitor cells was not detected in clonal cultures of bone marrow cells from mpl(-/-) mice. However, mpl(-/-) mice responded to IL-11 in vivo as evidenced by a rise in platelet count and an increase in spleen weight. Experiments were performed to address this paradox: administration of 5-fluorouracil with consequent "expansion" of early hematopoietic cells resulted in the appearance of IL-11-responsive cells in mpl(-/-) mice when assayed in in vitro cultures. CONCLUSIONS Thus, although mpl(-/-) mice have the capacity to produce IL-11-responsive progenitor cells, under steady state conditions their expansion is dependent on TPO. This is the first evidence that a specific subset of bone marrow progenitor/stem cells is altered in mpl(-/-) mice.
Collapse
Affiliation(s)
- C L Scott
- The Walter and Eliza Hall Institute of Medical Research, P.O. Royal Melbourne Hospital, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
86
|
Sloand E. Hematopoiesis in HIV infection: use of colony stimulating factors and cytokines. Cancer Treat Res 2001; 104:329-46. [PMID: 11191133 DOI: 10.1007/978-1-4615-1601-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
|
87
|
Fields KK, Crump M, Bence-Bruckler I, Bernstein S, Williams S, Frankel S, Miller A, Demetri G, Nabholtz JM, Cruickshank S, Lill M. Use of PEG-rHuMGDF in platelet engraftment after autologous stem cell transplantation. Bone Marrow Transplant 2000; 26:1083-8. [PMID: 11108307 DOI: 10.1038/sj.bmt.1702662] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This paper summarizes a pilot, sequential dose-escalation study of PEG-rHuMGDF in patients with advanced malignancies who had delayed platelet recovery after autologous stem cell transplantation (ASCT). Patients were randomized to receive either placebo (n = 11) or PEG-rHuMGDF at 5 (n = 9), 10 (n = 6), or 25 (n = 7) microg/kg/day by subcutaneous injection for 14 days and were monitored for 5 weeks. Across all treatment groups, eight patients had platelet recovery to > or = 20 x 10(9)/l by day 21. The proportion of patients achieving platelet recovery, the median number of days and units of platelet transfusions were similar for the placebo and the PEG-rHuMGDF groups. PEG-rHuMGDF was well tolerated at all dosages. The incidence rates of adverse events in all groups were similar. No deaths on study, no drug-related serious adverse events, and no development of neutralizing antibodies to MGDF occurred.
Collapse
Affiliation(s)
- K K Fields
- H Lee Moffitt Cancer Center, Tampa, FL 33617, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Scott CL, Robb L, Mansfield R, Alexander WS, Begley CG. Granulocyte-macrophage colony-stimulating factor is not responsible for residual thrombopoiesis in mpl null mice. Exp Hematol 2000; 28:1001-7. [PMID: 11008012 DOI: 10.1016/s0301-472x(00)00504-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To examine the role of granulocyte-macrophage colony-stimulating factor (GM-CSF) in thrombopoiesis. MATERIALS AND METHODS Thrombopoietin-unresponsi ve mice (mpl null mice), which have a profound reduction in platelets and mature megakaryocytes, were interbred with mice that do not respond to GM-CSF or interleukin 5 (betac null mice), and hematopoiesis was examined. In initial experiments on a mixed genetic background, double mutant mice (betac/mpl null mice) showed an unexpected amelioration of the thrombocytopenia seen in mpl null mice. Platelet counts were elevated approximately twofold in betac/mpl null mice compared with mpl null mice (mpl null 73+/-31; betac/mpl null 164+/-70; n = 10 to 29 mice per genotype, p<0.00001). This was associated with lessening of the deficit of megakaryocytes, progenitor cells, and colony-forming units spleen seen in mpl null mice. This amelioration of the mpl null phenotype in betac/mpl null mice on a mixed genetic background was highly statistically significant. To determine whether this amelioration of phenotype was solely the consequence of loss of betac signaling, progeny of a second intercross on a C57BL/6 background (B6betac/mpl null mice) were examined. When the resulting B6betac/mpl null mice were analyzed and compared with B6mpl null littermates, the increase in platelet count, hematopoietic progenitor cell number, and colony-forming units spleen number was no longer observed. CONCLUSION There was no additional effect seen as a result of loss of betac signaling. GM-CSF did not play a significant role in thrombopoiesis, even in combination with the absence of thrombopoietin signaling. These results highlight problems that can be encountered when studying introduced mutations in mice. They exemplify the importance of eliminating the influence of modifying genes when attributing biologic differences to specific introduced genetic alterations.
Collapse
Affiliation(s)
- C L Scott
- The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.
| | | | | | | | | |
Collapse
|
89
|
Sola MC, Del Vecchio A, Rimsza LM. Evaluation and treatment of thrombocytopenia in the neonatal intensive care unit. Clin Perinatol 2000; 27:655-79. [PMID: 10986634 DOI: 10.1016/s0095-5108(05)70044-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Thrombocytopenia is a very frequent problem among sick neonates, affecting up to 35% of all infants admitted to the NICU. Although multiple clinical conditions have been causally associated with neonatal thrombocytopenia, the cause of the thrombocytopenia is unclear in up to 60% of affected neonates. This article provides neonatologists with a practical approach to the thrombocytopenic neonate, with an emphasis on conditions that could be life-threatening or could have significant implications for further pregnancies. An overview of the current therapeutic modalities is also presented, including a discussion of the possible use of recombinant thrombopoietic cytokines to treat certain groups of thrombocytopenic neonates.
Collapse
Affiliation(s)
- M C Sola
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, USA.
| | | | | |
Collapse
|
90
|
Basser RL, Underhill C, Davis I, Green MD, Cebon J, Zalcberg J, MacMillan J, Cohen B, Marty J, Fox RM, Begley CG. Enhancement of platelet recovery after myelosuppressive chemotherapy by recombinant human megakaryocyte growth and development factor in patients with advanced cancer. J Clin Oncol 2000; 18:2852-61. [PMID: 10920133 DOI: 10.1200/jco.2000.18.15.2852] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To explore the influence of dose and schedule on the ability of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) to abrogate thrombocytopenia after multiple cycles of chemotherapy and to mobilize peripheral-blood progenitor cells (PBPC). PATIENTS AND METHODS In this open-label study, 68 patients with advanced cancer were randomized to receive PEG-rHuMGDF subcutaneously at different doses and durations before administration of carboplatin 600 mg/m(2), cyclophosphamide 1,200 mg/m(2), and filgrastim 5 microgram/kg/d. PEG-rHuMGDF was not given after the first cycle of chemotherapy but was given after the second and subsequent cycles. Chemotherapy was given every 28 days for up to six cycles. RESULTS In patients who received the same dose of chemotherapy for at least two cycles, the platelet nadir was significantly higher (47.5 x 10(9)/L v 35.5 x 10(9)/L; P =.003) and duration of grade 3 or 4 thrombocytopenia significantly shorter (0 v 3 days; P =.004) when PEG-rHuMGDF was administered after chemotherapy. There was no evidence of an effect of PEG-rHuMGDF when it was given before chemotherapy. Platelet recovery after the first cycle of chemotherapy was no different for different PEG-rHuMGDF regimens, and there was no difference between patients treated with PEG-rHuMGDF and historical controls treated with identical chemotherapy. There was a modest dose-related increase in progenitor cell levels after administration of PEG-rHuMGDF alone. Peak levels of PBPC occurred later in cycle 2 than in cycle 1 but were not different in magnitude. CONCLUSION PEG-rHuMGDF abrogated severe thrombocytopenia after dose-intensive chemotherapy. However, it had only a modest effect on progenitor cell levels and did not enhance progenitor cell mobilization after chemotherapy and filgrastim.
Collapse
Affiliation(s)
- R L Basser
- Centre for Developmental Cancer Therapeutics, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
McNiece IK. New cytokines and their clinical application. Cancer Treat Res 2000; 101:389-405. [PMID: 10800659 DOI: 10.1007/978-1-4615-4987-1_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Affiliation(s)
- I K McNiece
- Bone Marrow Transplant Unit, University of Colorado Health Sciences Center, Denver 80262, USA
| |
Collapse
|
92
|
Abstract
C-mpl ligand or thrombopoietin (Tpo) is increasingly recognised as the major regulator of platelet homeostasis in humans. Relatively little is known about Tpo in the fetus and neonate but no evidence has yet been found to suggest any fundamental difference in Tpo structure, function and regulation in the fetus and neonate compared to older age groups. Tpo mRNA transcripts have been detected in the fetus as early as 6 weeks post conception and the liver appears to be the main site of Tpo production in both the fetus and neonate. The vast majority of healthy newborns have detectable levels of circulating Tpo and raised Tpo levels are commonly, but not consistently, found in thrombocytopenic neonates. In adults receptor binding and subsequent metabolism of Tpo is proposed as the main method of regulation of the circulating Tpo level. Preliminary studies in neonates showing increased Tpo levels most often during thrombocytopenia accompanied by reduced megakaryocytopoiesis supports this concept. In addition to this demonstrable fetal and neonatal endogenous Tpo production megakaryocyte progenitor and precursor cells from the fetus and from preterm and term newborns proliferate and differentiate extensively in-vitro in response to exogenous Tpo. Furthermore a recent study has shown a marked rise in platelet count in newborn rhesus monkeys administered one form of recombinant Tpo. Although these studies remain at an early stage together these findings strongly suggest that, as in adults, Tpo is the major regulator of platelet homeostasis in the fetus and neonate. Thrombocytopenia is common in sick neonates and progress in understanding this important clinical problem is likely to be greatly enhanced by the current and future research into Tpo production, function and regulation in the healthy and thrombocytopenic fetus and neonate.
Collapse
Affiliation(s)
- N A Murray
- Department of Neonatal Medicine, Imperial College School of Medicine, Hammersmith Campus, Hammersmith Hospital, Du Cane Road, W12 ONN, London, UK.
| | | | | |
Collapse
|
93
|
Bolwell B, Vredenburgh J, Overmoyer B, Gilbert C, Chap L, Menchaca DM, Cruickshank S, Glaspy J. Phase 1 study of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) in breast cancer patients after autologous peripheral blood progenitor cell (PBPC) transplantation. Bone Marrow Transplant 2000; 26:141-5. [PMID: 10918423 DOI: 10.1038/sj.bmt.1702465] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Forty-seven patients with stage II, III, or IV breast cancer undergoing autologous peripheral blood progenitor cell (PBPC) transplantation were randomized to placebo (n = 13) or to one of five sequential dose cohorts of pegylated (PEG) recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) (1.0, 2.5, 5.0, 7.5, or 10.0 microg/kg/day) (n= 34). Blinded study drug was started on the day of transplantation and was continued until the platelet count was > or =100 x 109/l or a maximum of 21 days. PBPCs were mobilized with filgrastim (r-metHuG-CSF) and all patients received filgrastim starting on day +2 after transplantation. The nadir platelet count was not affected by treatment. The median time to platelet recovery was 11 and 12 days for the placebo and combined PEG-rHuMGDF groups, respectively. No trends in adverse events suggested dose- or treatment-related toxicity. Two patients withdrew from the study because of an adverse event (allergic reaction in the 7.5 microg/kg group) probably related to study drug, and veno-occlusive disease (VOD) (in the 5 microg/kg group) which was felt not to be related to study drug by the investigator. No patients developed neutralizing antibodies to MGDF. Day +21 and day +28 platelet counts were higher in the group receiving PEG-rHuMGDF (246 vs 148 x 109/l and 299 vs 145 x 109/l, respectively; both P < 0. 05). PEG-rHuMGDF up to 10 microg/kg/day was well tolerated. In this study, there was no effect of study drug on initial platelet engraftment at the doses studied. However, the efficacy of other doses is unknown.
Collapse
Affiliation(s)
- B Bolwell
- The Cleveland Clinic Foundation, OH 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|
94
|
Goodnough LT, Kuter D, McCullough J, Brecher ME. Apheresis platelets: emerging issues related to donor platelet count, apheresis platelet yield, and platelet transfusion dose. J Clin Apher 2000; 13:114-9. [PMID: 9828021 DOI: 10.1002/(sici)1098-1101(1998)13:3<114::aid-jca4>3.0.co;2-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Emerging issues in stimulating apheresis platelet donors with platelet growth factors, the relative costs of apheresis and random donor platelet concentrates, optimal platelet transfusion dose, and leucoreduction of platelet products have caused renewed debate regarding apheresis products vs. random, pooled concentrates. The future role of apheresis products in platelet transfusion therapy will in large part be determined by costs, which are increasingly recognized to be influenced by donor platelet count, apheresis yield, and platelet transfusion dose.
Collapse
Affiliation(s)
- L T Goodnough
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.
| | | | | | | |
Collapse
|
95
|
Español I, Pujol-Moix N. [Thrombopoietin: its discovery and clinical perspectives]. Med Clin (Barc) 2000; 114:511-6. [PMID: 10846658 DOI: 10.1016/s0025-7753(00)71347-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- I Español
- Departament d'Hematologia, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona
| | | |
Collapse
|
96
|
Abstract
Since the purification of thrombopoietin 6 years ago, c-Mpl ligands such as recombinant human thrombopoietin (rhTPO) and pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF) have undergone extensive clinical investigation. Both recombinant forms have been shown to reduce the thrombocytopenia associated with nonmyeloablative chemotherapy. Several areas of research have been identified for further clinical development of c-Mpl ligands. One future direction is to continue to explore the intravenous route of administration of rhTPO and PEG-rHuMGDF, as well as fusion proteins of interleukin-3-thrombopoietin and thrombopoietin peptide mimetics, which may be as potent as thrombopoietin, but may lack antigenicity. Another focus would be on the use of these molecules in treating non-chemotherapy-induced thrombocytopenia associated with myelodysplastic syndrome (MDS), idiopathic thrombocytopenic purpura (ITP), human immunodeficiency virus (HIV)-related ITP, and liver disease. Additionally, c-Mpl ligands may have a role in improving apheresis yields when administered to normal platelet donors. Considerable data demonstrate the effectiveness of PEG-rHuMGDF in raising the platelet yields in apheresis donors. In the past few years, investigation into the use of thrombopoietin for ex vivo expansion of pluripotent stem cells has been extensive. Last, thrombopoietin may serve as a radioprotectant by preventing radiation-induced apoptosis of pluripotent stem cells. In the coming years, the clinical role of rhTPO, PEG-rHuMGDF, and related molecules such as the thrombopoietin peptide mimetics will probably be established for both chemotherapeutic and nonchemotherapeutic indications.
Collapse
Affiliation(s)
- D J Kuter
- Hematology/Oncology Unit, Massachusetts General Hospital, Harvard Medical School, Boston 02114, USA
| |
Collapse
|
97
|
Abstract
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, IL-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 justify testing in randomized clinical 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 megakaryocyte 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 rhTPO, the future may see the development of novel genetically engineered, high-affinity cytokine receptor agonists and c-Mpl ligand mimetic peptides.
Collapse
Affiliation(s)
- G D Demetri
- Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, MA 02115-6084, USA
| |
Collapse
|
98
|
Abstract
The search for a thrombopoietic agent has resulted in the identification of numerous cytokines and growth factors with thrombopoietic activity. However, with the exception of interleukin (IL)-11 and thrombopoietin (TPO), the megakaryopoietic activity of most of these molecules has not produced clearly identifiable clinical benefits. Despite the relatively modest effect of IL-11 on megakaryocyte and platelet production in vitro and in vivo, it does reduce the need for platelet transfusions in specialized clinical settings. In contrast, the c-Mpl ligand TPO has been shown to be a potent stimulator of megakaryocyte and platelet production both in vitro and in vivo. Clinical studies are being conducted with two different preparations of the c-Mpl ligand: recombinant human thrombopoietin (rhTPO) and pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF). A recombinant form of the complete human molecule, rhTPO is glycosylated and produced in mammalian cells. PEG-rHuMGDF consists of only the receptor-binding domain linked to a polyethylene glycol (PEG) moiety and is generated in Escherichia coil. Although c-Mpl ligands are still being evaluated, preliminary evidence indicates that these molecules can elevate platelet counts and may be useful in a range of clinical contexts. This report discusses aspects of the biology behind the clinical actions of IL-11 and the c-Mpl ligands.
Collapse
Affiliation(s)
- C G Begley
- Centre for Developmental Cancer Therapeutics, Parkville, Victoria, Australia
| | | |
Collapse
|
99
|
Yoshimura C, Nomura S, Katsura K, Yamaguchi K, Fukuhara S. Thrombopoietin Levels in Patients Undergoing Autologous Peripheral Blood Stem Cell Transplantation. Vox Sang 2000. [DOI: 10.1046/j.1423-0410.2000.7820106.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
100
|
Tokunaga Y, Miyamoto T, Gondo H, Okamura T, Niho Y. Effect of thrombopoietin on acute myelogenous leukemia blasts. Leuk Lymphoma 2000; 37:27-37. [PMID: 10721767 DOI: 10.3109/10428190009057626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It has been shown that the expression of c-mp1, which is a specific receptor for thrombopoietin (TPO), is restricted to the surface of megakaryocytes, platelets, human CD34+ progenitor cells and human erythroid/megakaryocytic leukemic cell lines. Recently, however, it has been reported that some acute myelogenous leukemia (AML) blasts expressed c-mp1 on their cell surface and proliferated in response to TPO. We therefore investigated the effect of thrombopoietin on the growth of leukemic blasts from patients with CD7-positive acute myelogenous leukemia (AML), which is a distinct biological and clinical subtype of AML. Significant growth responses of leukemic blasts to TPO were seen in 10/10 CD7+ and 7/20 CD7- AML cases using 3H-thymidine incorporation, while synergistic stimulatory effects of TPO with stem cell factor (SCF), interleukin-3 (IL-3), granulocyte colony-stimulating factor, and granulocyte-macrophage colony-stimulating factor were observed in both groups. In a leukemic blast colony assay, significant growth response to TPO was observed in 5/6 CD7+ and 4/17 CD7- AML cases examined. Furthermore, the expression of c-mp1 seemed to be higher in CD7+ AML cases than in CD7- cases, suggesting a relationship between the expression of c-mp1 and the proliferative response to TPO. These findings imply that CD7+ leukemic blasts express functional TPO receptors and proliferate in response to TPO. Thus CD7 expression on AML blasts may indicate the involvement of leukemic progenitors at an early stage of multipotent hemopoietic stem cells. In this review, we discuss the effect of TPO on AML blasts, especially in CD7+ AML cases.
Collapse
Affiliation(s)
- Y Tokunaga
- First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | |
Collapse
|