Skip to main content
Log in

Filgrastim

A Review of its Pharmacological Properties and Therapeutic Efficacy in Neutropenia

  • Drug Evaluation
  • Published:
Drugs Aims and scope Submit manuscript

Summary

Synopsis

Filgrastim, a recombinant human granulocyte colony-stimulating factor (G-CSF), has identical biological activity to that of endogenous human G-CSF, but differs in that it contains an N-terminal methionine residue and is not glycosylated. It principally stimulates activation, proliferation and differentiation of neutrophil progenitor cells and has been evaluated in the treatment of patients with various neutropenic conditions, both iatrogenic and disease-related.

Two comparative studies have demonstrated that prophylactic administration of filgrastim 230 μg/m2/day significantly reduces the incidence, duration and severity of neutropenia in patients with previously untreated small-cell lung cancer receiving standard-dose chemotherapy with CDE (cyclophosphamide, doxorubicin plus etoposide). Concomitant with the amelioration of neutropenia, the incidence of febrile neutropenia was significantly reduced by 50% and there were 35 and 50% decreases in hospitalisation rates and intravenous antibiotic requirements.

Since not all patients receiving standard-dose chemotherapy are at risk of infectious complications, prophylactic filgrastim use may be reserved for those patients who have developed febrile neutropenia during a previous cycle of the same regimen. This strategy may prove less costly, although potential savings must be weighed against a greater risk of patient morbidity and reduced quality of life. When combined with standard intravenous antibiotic therapy, filgrastim further decreases morbidity in patients with established febrile neutropenia and may have a positive impact on overall treatment costs by shortening the length of hospitalisation.

Attention is focused on the use of haematopoietic growth factors to support dose-intensification of chemotherapy with a view to improving treatment outcomes in patients with chemo-responsive tumours. Filgrastim, used alone, permits modest increases in dose-intensity and/or dose-escalation of some standard-dose chemotherapy regimens. Moreover, the drug has proven useful as an adjunct to myeloablative chemotherapy followed by stem cell rescue with autologous bone marrow transplantation and/or peripheral blood progenitor cells. However, the impact of these dose-intensification approaches on survival remains to be determined in well-controlled clinical studies.

Filgrastim is effective in increasing the neutrophil count and decreasing morbidity in patients with severe chronic neutropenia, including Kostmann’s syndrome, and in idiopathic and cyclic neutropenia. In addition, filgrastim has accelerated neutrophil recovery in patients with idiosyncratic drug-induced agranulocytosis.

Available data indicate that filgrastim is generally well tolerated. The most frequent adverse reaction is mild to moderate medullary bone pain, reported by approximately 20% of patients, although this can generally be controlled using simple analgesics without the need to discontinue treatment.

In summary, filgrastim should be regarded as a first-line therapy in patients with severe chronic neutropenia and, when used judiciously, a valuable adjuvant to cytotoxic cancer chemotherapy in patients with nonmyelogenous malignancies. Further clinical experience should help define a wider role for the drug in the treatment and management of other conditions associated with neutropenia, including acquired immunodeficiency syndrome.

Pharmacological Properties

Filgrastim (recombinant methionyl human granulocyte colony-stimulating factor) is a haematopoietic growth factor which acts primarily to stimulate proliferation and differentiation of committed progenitor cells of the granulocyte-neutrophil lineage into functionally mature neutrophils. The neutrophilia induced by filgrastim is characterised by a dose-dependent ‘left-shift’ towards progenitor cells; mature neutrophils show normal or enhanced function in vitro after exposure to filgrastim.

Filgrastim causes a transient decrease in the absolute neutrophil count (ANC) within the first hour after administration, followed by a rapid increase within 4 to 5 hours. The increase in ANC was 2- to 12-fold over pretreatment values after subcutaneous administration of filgrastim 1 to 60 μg/kg and was dose-proportional. Neutrophil levels decrease exponentially to normal levels generally within 2 days of the last dose.

In cancer patients receiving chemotherapy, administration of filgrastim shortened the time to occurrence of the neutrophil nadir and dose-proportionally decreased the duration and severity of neutropenia. However, the neutrophil response to conventional dosages of filgrastim may vary, depending on the previous level of chemotherapy or radiotherapy.

Filgrastim 1 to 60 μg/kg does not affect eosinophil or basophil counts but causes a minor dose-dependent increase in monocyte levels at higher doses (10 to 60 μg/kg). Minor increases in lymphocytes may also occur at high doses, probably by an indirect mechanism.

Peak plasma concentrations of filgrastim are dose-proportional and occurred 2 to 8 hours after subcutaneous administration in children with severe chronic neutropenia. Filgrastim clearance appears to be dependent on the neutrophil count; clearance increased from 0.024 to 0.044 L/h (0.40 to 0.74 ml/min) over 14 days as the ANC increased in children with severe chronic neutropenia. Similarly, the elimination half-life decreased from 4.7 to <2 hours as the ANC increased. The mechanism by which filgrastim is metabolised and excreted is not known.

Therapeutic Efficacy

Filgrastim has been widely evaluated as an adjunct to chemotherapy in the treatment of nonmyelogenous cancers, although its use in myelodysplastic syndromes and leukaemia should be regarded as experimental pending the availability of further clinical data.

In 2 placebo-controlled studies, prophylactic administration of filgrastim 230 μg/m2/day for up to 14 days significantly reduced the incidence, severity and duration of neutropenia in patients with previously untreated small-cell lung cancer receiving standard-dose chemotherapy with CDE (cyclophosphamide, doxorubicin plus etoposide). As a result, there was a significant 50% reduction in the incidence of febrile neutropenia and this was associated with 35 and 50% decreases in the use of measured healthcare resources (hospitalisations and intravenous antibiotic requirements). In other randomised studies, filgrastim further reduced morbidity when combined with prophylactic antimicrobial and antifungal therapy and standard antibiotic treatment in patients with established febrile neutropenia.

Various pharmacoeconomic analyses of filgrastim therapy have been reported. Generally, these show that the cost of providing prophylactic filgrastim therapy can at least be partially recouped through savings in hospitalisation costs. However, careful patient selection is required to maximise the potential cost-benefits associated with prophylactic filgrastim use. In contrast, therapeutic administration to patients with febrile neutropenia receiving standard antibiotic treatment has reduced overall treatment costs by 40% compared with placebo, although further studies are required to confirm this promising result.

When administered to patients receiving high-dose chemotherapy followed by bone marrow transplantation (BMT), filgrastim accelerates the recovery of adequate neutrophil levels. Compared with historical controls, filgrastim tends to reduce the severity and duration of infection-related complications in patients receiving autologous bone marrow transplantation (ABMT), reflected in fewer days with febrile neutropenia as well as significant reductions in antibiotic use and hospitalisation times. However, significant effects have varied between different centres and confirmation of the clinical benefits of filgrastim is required in detailed reports of randomised studies.

The use of peripheral blood progenitor cells (PBPCs) administered in conjunction with, or even as an alternative to, BMT is now being investigated. Data from nonrandomised studies indicate that PBPC infusion accelerates haematopoietic reconstitution after high-dose chemotherapy and ABMT, with significant reductions in hospital stay, antibiotic use and transfusion requirements. Mobilisation of PBPCs with filgrastim has been successfully integrated into induction- or salvage-chemotherapy regimens, although the response may vary depending on the previous level of chemotherapy.

Filgrastim 3.4 to 11.5 μg/kg/day was effective in reducing the incidence and duration of infection in patients with severe chronic neutropenia, including idiopathic, cyclic and congenital neutropenias. An increase in ANC to >1.5 × 109/L was seen in 90% of patients. The data available suggest that filgrastim is more effective than granulocyte-macrophage colony-stimulating factor in these patients.

Preliminary findings suggest that coadministration of filgrastim with the antiviral drugs zidovudine or ganciclovir is effective in attenuating the leucopenia associated with these agents, thus permitting continued therapy in disorders such as acquired immunodeficiency syndrome (AIDS). Filgrastim may also be useful in accelerating neutrophil recovery in patients with idiosyncratic drug-induced agranulocytosis.

Tolerability

Filgrastim is generally well tolerated. The most common adverse reaction is mild to moderate medullary bone pain, which is normally controlled by simple analgesics and generally resolves on continued treatment. In placebo-controlled trials, bone pain occurred in 20% of patients with cancer receiving chemotherapy and filgrastim.

Minor increases in plasma levels of leucocyte alkaline phosphatase, lactate dehydrogenase and uric acid are frequent with filgrastim, and are possibly related to the increase in neutrophils. Asymptomatic splenomegaly occurs in about 25% of patients receiving long term treatment with filgrastim for severe chronic neutropenia.

Rare adverse events occurring during filgrastim therapy include vasculitis, acute neutrophilic dermatosis and possible anaphylactoid reactions. Filgrastim has not been reported to cause increased eosinophil levels. G-CSF has rarely been reported to cause capillary leak syndrome.

Dosage and Administration

Filgrastim can be administered by subcutaneous injection or infusion, or intravenous infusion. The subcutaneous route allows the drug to be administered on an outpatient basis where this is appropriate. An initial starting dose of 5 μg/kg/day is recommended for the treatment of chemotherapy-induced neutropenia and idiopathic and cyclic neutropenias. A higher starting dose of 10 μg/kg/day is recommended in patients receiving high-dose chemotherapy followed by bone marrow transplant, and in patients with congenital neutropenias such as Kostmann’s syndrome.

Caution should be exercised when administering filgrastim to patients with myeloid malignancies or myelodysplastic syndromes because of the possibility of filgrastim promoting tumour growth or progression to malignancy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

References

  1. Ariad S, Geffen D. Role of colony-stimulating factors during high dosage chemotherapy. Clin Immunother 1994; 1: 449–59

    Google Scholar 

  2. Hollingshead LM, Goa KL. Recombinant granulocyte colonystimulating factor (rG-CSF). A review of its pharmacological properties and prospective role in neutropenic conditions. Drugs 1991; 42: 300–30

    CAS  PubMed  Google Scholar 

  3. Grant SM, Heel RC. Recombinant granulocyte-macrophage colony-stimulating factor (rGM-CSF): a review of its pharmacological properties and prospective role in the management of myelosuppression. Drugs 1992; 43: 516–60

    CAS  PubMed  Google Scholar 

  4. Faulds D, Sorkin EM. Epoetin (recombinant human erythropoietin): a review of is pharmacodynamic and pharmacokinetic properties and therapeutic potential in anaemia and the stimulation of erythropoiesis. Drugs 1989; 38: 863–99

    CAS  PubMed  Google Scholar 

  5. Souza LM, Boone TC, Gabrilove J, et al. Recombinant human granulocyte colony-stimulating factor: effects on normal and leukemic myeloid cells. Science 1986; 232: 61–5

    CAS  PubMed  Google Scholar 

  6. Lord BI, Bronchud MH, Owens S, et al. The kinetics of human granulopoiesis following treatment with granulocyte colony-stimulating factor in vivo. Proc Natl Acad Sci USA 1989; 86: 9499–503

    CAS  PubMed  Google Scholar 

  7. Bronchud MH, Potter MR, Morgenstern G, et al. In vitro and in vivo analysis of the effects of recombinant human granulocyte colony-stimulating factor in patients. Br J Cancer 1988; 58: 64–9

    CAS  PubMed  Google Scholar 

  8. Itoh Y, Kuratsuji T, Tsunawaki S, et al. In vivo effects of recombinant human granulocyte colony-stimulating factor on normal neutrophil function and membrane effector molecule expression. Int J Hematol 1991; 54: 463–9

    CAS  PubMed  Google Scholar 

  9. Gabrilove JL, Jakubowski A, Fain K, et al. Phase I study of granulocyte colony-stimulating factor in patients with transitional cell carcinoma of the urothelium. J Clin Invest 1988; 82: 1454–61

    CAS  PubMed  Google Scholar 

  10. Lindermann A, Herrmann F, Oster W, et al. Hematologic effects of recombinant human granulocyte colony-stimulating factor in patients with malignancy. Blood 1989; 74: 2644–51

    Google Scholar 

  11. Morstyn G, Campbell L, Souza LM, et al. Effect of granulocyte colony stimulating factor on neutropenia induced by cytotoxic chemotherapy. Lancet 1988; 1: 667–72

    CAS  PubMed  Google Scholar 

  12. Morstyn G, Campbell L, Lieschke G, et al. Treatment of chemotherapy-induced neutropenia by subcutaneously administered granulocyte colony-stimulating factor with optimization of dose and duration of therapy. J Clin Oncol 1989; 7: 1554–62

    CAS  PubMed  Google Scholar 

  13. Di Leo A, Bajetta E, Nolè F, et al. The intramuscular administration of granulocyte colony-stimulating factor as an adjunct to chemotherapy in pretreated ovarian cancer patients: an Italian Trials in Medical Oncology (ITMO) Group pilot study. Br J Cancer 1993; 69: 961–6

    Google Scholar 

  14. Schmitz LL, McClure JS, Litz CE, et al. Morphologic and quantitative changes in blood and marrow cells following growth factor therapy. Am J Clin Pathol 1994; 101: 67–75

    CAS  PubMed  Google Scholar 

  15. Eisner J, Roesler J, Emmendörffer A, et al. Altered function and surface marker expression of neutrophils induced by rhGCSF treatment in severe congenital neutropenia. Eur J Haematol 1992; 48: 10–9

    Google Scholar 

  16. Roesler J, Emmendörffer A, Eisner J, et al. In vitro functions of neutrophils induced by treatment with rhG-CSF in severe congenital neutropenia. Eur J Haematol 1991; 46: 112–8

    CAS  PubMed  Google Scholar 

  17. McCawley LJ, Korchak HM, Douglas SD, et al. In vitro and in vivo effects of granulocyte colony-stimulating factor on neutrophils in glycogen storage disease type IB: granulocyte colony-stimulating factor therapy corrects the neutropenia and the defects in respiratory burst activity and Ca2+ mobilization. Pediatr Res 1994; 35: 84–90

    CAS  PubMed  Google Scholar 

  18. Ohsaka A, Kitagawa S, Yuo A, et al. Effects of granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor on respiratory burst activity of neutrophils in patients with myelodysplastic syndromes. Clin Exp Immunol 1993; 91: 308–13

    CAS  PubMed  Google Scholar 

  19. Diihrsen U, Villeval J-L, Boyd J, et al. Effects of recombinant human granulocyte colony-stimulating factor on hematopoietic progenitor cells in cancer patients. Blood 1988; 72: 2074–81

    Google Scholar 

  20. Schwinger W, Mache C, Urban C, et al. Single dose of filgrastim (rhG-CSF) increases the number of hematopoietic progenitors in the peripheral blood of adult volunteers. Bone Marrow Transplant 1993; 11: 489–92

    CAS  PubMed  Google Scholar 

  21. Dreger P, Grelle K, Eckstein V, et al. Granulocyte-colony-stimulating factor induces inceased serum levels of soluble inter-leukin 2 receptors preceding engraftment in autologous bone marrow transplantation. Br J Haematol 1993; 83: 7–13

    CAS  PubMed  Google Scholar 

  22. Berdel WE, Danhauser-Riedl S, Steinhauser G, et al. Various human hematopoietic growth factors (interleukin-3, GMCSF, G-CSF) stimulate clonal growth of nonhematopoietic tumor cells. Blood 1989; 73: 80–3

    CAS  PubMed  Google Scholar 

  23. Connor JP, Squatrito RC, Terrell KL, et al. In vitro growth effects of colony-stimulating factors in ovarian cancer. Gynecol Oncol 1994; 52: 347–52

    CAS  PubMed  Google Scholar 

  24. Segawa K, Suhara Y, Ueno Y, et al. Effect of co-administration of granulocyte colony-stimulating factor on interferon therapy. Jpn J Cancer Res 1991; 82: 346–50

    CAS  PubMed  Google Scholar 

  25. Layton JE, Hockman H, Sheridan WP, et al. Evidence for a novel in vitro control mechanism of granulopoiesis: mature cell-related control of a regulatory growth factor. Blood 1989; 74: 1301–7

    Google Scholar 

  26. Eguchi K, Shinkai T, Sasaki Y, et al. Subcutaneous administration of recombinant human granulocyte colony-stimulating factor (KRN8601) in intensive chemotherapy for patients with advanced lung cancer. Jpn J Cancer Res 1990; 81: 1168–74

    CAS  PubMed  Google Scholar 

  27. Kearns CM, Wang WC, Stute N, et al. Disposition of recombinant human granulocyte colony-stimulating factor in children with severe chronic neutropenia. J Pediatr 1993; 123: 471–9

    CAS  PubMed  Google Scholar 

  28. Stute N, Santana VM, Rodman JH, et al. Pharmacokinetics of subcutaneous recombinant human granulocyte colony-stimulating factor in children. Blood 1992; 79: 2849–54

    CAS  PubMed  Google Scholar 

  29. Filgrastim product monograph. Amgen Inc. and Roche Products Ltd, UK, 1994

  30. Lieschke GJ, Burgess AW. Granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor (second of two parts). N Engl J Med 1992; 327: 99–106

    CAS  PubMed  Google Scholar 

  31. Bronchud MH, Scarffe JH, Thatcher N, et al. Phase I/II study of recombinant human granulocyte colony-stimulating factor in patients receiving intensive chemotherapy for small cell lung cancer. Br J Cancer 1987; 56: 809–13

    CAS  PubMed  Google Scholar 

  32. Gabrilove JL, Jakubowski A, Scher H, et al. Effect of granulocyte colony-stimulating factor on neutropenia and associated morbidity due to chemotherapy for transitional-cell carcinoma of the urothelium. N Engl J Med 1988; 318: 1414–22

    CAS  PubMed  Google Scholar 

  33. Crawford J, Ozer H, Stoller R, et al. Reduction by granulocyte colony-stimulating factor of fever and neutropenia induced by chemotherapy in patients with small-cell lung cancer. N Engl J Med 1991; 325: 164–70

    CAS  PubMed  Google Scholar 

  34. Trillet-Lenoir V, Green J, Manegold C, et al. Recombinant granulocyte colony stimulating factor reduces the infectious complications of cytotoxic chemotherapy. Eur J Cancer 1993; 29A: 319–24

    CAS  PubMed  Google Scholar 

  35. Miller LL. Current status of G-CSF in support of chemotherapy and radiotherapy. Oncology 1993; 7: 67–78

    CAS  PubMed  Google Scholar 

  36. Pettengell R, Gurney H, Radford JA, et al. Granulocyte colonystimulating factor to prevent dose-limiting neutropenia in non-Hodgkin’s lymphoma: a randomized controlled trial. Blood 1992; 80: 1430–6

    CAS  PubMed  Google Scholar 

  37. Masuda N, Fukuoka M, Furuse K, et al. CODE chemotherapy with or without recombinant human granulocyte colony-stimulating factor in extensive-stage small cell lung cancer. Oncology 1992; 49 Suppl. 1: 19–24

    PubMed  Google Scholar 

  38. Dunlop DJ, Anderson S, Paul J, et al. Filgrastim as an adjunct to combination chemotherapy in Hodgkin’s disease [abstract]. Br J Cancer 1994 Mar; 69 Suppl. 21:11

    Google Scholar 

  39. Maher D, Green M, Bishop J, et al. Randomized, placebo-controlled trial of filgrastim in patients with febrile neutropenia following chemotherapy [abstract no. 1498]. Proc Am Soc Clin Oncol 1993; 12: 434

    Google Scholar 

  40. Mayordomo JI, Rivera F, Diaz-Puente MT, et al. Decreasing morbidity and cost of treating febrile neutropenia by adding G-CSF and GM-CSF to standard antibiotic therapy; results of a randomized trial [abstract no. 1510]. Proc Am Soc Clin Oncol 1993; 12: 437

    Google Scholar 

  41. Faulds D, Lewis NJW, Milne RJ. Recombinant granulocyte colony-stimulating factor (rG-CSF): pharmacoeconomic considerations in chemotherapy-induced neutropenia. PharmacoEconomics 1992; 1: 231–49

    CAS  PubMed  Google Scholar 

  42. Glaspy JA, Bleecker G, Crawford J, et al. The impact of therapy with filgrastim (recombinant granulocyte colony-stimulating factor) on the health care costs associated with cancer chemotherapy. Eur J Cancer A 1993 Dec; 29A Suppl. 7: S23–30

    PubMed  Google Scholar 

  43. Nichols CR, Fox EP, Roth BJ, et al. Incidence of neutropenic fever in patients treated with standard-dose combination chemotherapy for small-cell lung cancer and the cost impact of treatment with granulocyte colony-stimulating factor. J Clin Oncol 1994; 12: 1245–50

    CAS  PubMed  Google Scholar 

  44. Tirelli U, Errante D, Tavio P, et al. Treatment of HIV-related non-Hodgkin’s lymphoma (NHL) with chemotherapy (CT) and G-CSF: reduction in the days of hospitalisation and toxicity with concomitant overall reduction in the cost [abstract]. Proc Am Soc Clin Oncol 1993; 12: 53

    Google Scholar 

  45. Zagonel V, Babare R, Merola MC, et al. Cost-benefit of granulocyte colony-stimulating factor administration in older patients with non-Hodgkin’s lymphoma treated with combination chemotherapy. Ann Oncol 1994; 5 Suppl. 2: 127–32

    PubMed  Google Scholar 

  46. Bronchud M. Can hematopoietic growth factors be used to improve the success of cytotoxic chemotherapy?. Anti Cancer Drugs 1993; 4: 127–39

    CAS  PubMed  Google Scholar 

  47. Budman DR, Wood W, Henderson IC, et al. Initial findings of CALGB 8541: a dose and dose intensity trial of cyclophosphamide (C), doxorubicin (A) and 5-fluorouracil (F) as adjuvant treatment of stage II, node+, female breast cancer [abstract no. 29]. Proc Am Soc Clin Oncol 1992; 11: 51

    Google Scholar 

  48. Bronchud MH, Howell A, Crowther D, et al. The use of granulocyte colony-stimulating factor to increase the intensity of treatment with doxorubicin in patients with ovarian cancer. Br J Cancer 1989; 60: 121–5

    CAS  PubMed  Google Scholar 

  49. Seidman AD, Scher HI, Gabrilove JL, et al. Dose-intensification of MVAC with recombinant granulocyte colony-stimulating factor as initial therapy in advanced urothelial cancer. J Clin Oncol 1993; 11: 408–14

    CAS  PubMed  Google Scholar 

  50. Steinke B, Münz A, Manegold C, et al. Treatment intensification with r-metHuG-CSF in high-grade malignant non-Hodgkin’s lymphomas. Onkologie 1994; 17: 248–53

    Google Scholar 

  51. Filgrastim package insert. Amgen Inc, USA, California, 1994.

  52. Schmitz N, Dreger P, Zander A, et al. A randomized controlled multicentre study of granulocyte colony stimulating factor (filgrastim) in patients with Hodgkin’s lymphoma undergoing autologous bone marrow transplantation [abstract]. Blood 1993; 82 Suppl. 1:568

    Google Scholar 

  53. Blaise D, Vernant JP, Fiere D, et al. A randomised, controlled, multicenter trial of recombinant human granulocyte colony stimulating factor (filgrastim) in patients treated by bone marrow transplantation (BMT) with total body irradiation (TBI) for acute lymphoblastic leukaemia (ALL) or lymphoblastic lymphoma (LL) [abstract 248a]. Blood 1992; 80: 982

    Google Scholar 

  54. Sheridan WP, Morstyn G, Wolf M, et al. Granulocyte colony-stimulating factor and neutrophil recovery after high-dose chemotherapy and autologous bone marrow transplantation. Lancet 1989; 1: 891–5

    Google Scholar 

  55. Taylor KM, Jagannath S, Spitzer G, et al. Recombinant human granulocyte colony-stimulating factor hastens granulocyte recovery after high-dose chemotherapy and autologous bone marrow transplantation in Hodgkin’s disease. J Clin Oncol 1989; 7: 1791–9

    CAS  PubMed  Google Scholar 

  56. Kennedy MJ, Davis J, Passos-Coelho J, et al. Administration of human recombinant granulocyte colony-stimulating factor (filgrastim) accelerates granulocyte recovery following high-dose chemotherapy and autologous marrow transplantation with 4-hydroperoxycyclophosphamide-purged marrow in women with metastatic breast cancer. Cancer Res 1993; 53: 5424–8

    CAS  PubMed  Google Scholar 

  57. Peters WP, Rosner G, Ross M, et al. Comparative effects of granulocyte-macrophage colony stimulating factor (GM-CSF) and granulocyte colony- stimulating factor (G-CSF) on priming peripheral blood progenitor cells for use with autologous bone marrow after high-dose chemotherapy. Blood 1993; 81: 1709–19

    CAS  PubMed  Google Scholar 

  58. Sheridan WP, Begley CG, Juttner CA, et al. Effect of peripheral-blood progenitor cells mobilised by filgrastim (G-CSF) on platelet recovery after high-dose chemotherapy. Lancet 1992; 339: 640–4

    CAS  PubMed  Google Scholar 

  59. Golde DW. Production, distribution and fate of neutrophils. In: Williams WJ, et al., editors. Hematology. 3rd ed. Singapore: McGraw-Hill Book Co., 1993: 759–65

    Google Scholar 

  60. Russell NH, Hunter A, Rogers S, et al. Peripheral blood stem cells as an alternative to marrow for allogeneic transplantation [letter]. Lancet 1993; 341: 1482

    CAS  PubMed  Google Scholar 

  61. Weaver CH, Buckner CD, Longin K, et al. Syngeneic transplantation with peripheral blood mononuclear cells collected after the administration of recombinant human granulocyte colony-stimulating factor. Blood 1993; 82: 1981–4

    CAS  PubMed  Google Scholar 

  62. Bensinger WI, Price TH, Dale DC, et al. The effects of daily recombinant human granulocyte colony-stimulating factor administration on normal granulocyte donors undergoing leukapheresis. Blood 1993; 7: 1883–8

    Google Scholar 

  63. Bensinger W, Singer J, Appelbaum K, et al. Autologous transplantation with peripheral blood mononuclear cells collected after administration of recombinant granulocyte stimulating factor. Blood 1993; 81: 3158–63

    CAS  PubMed  Google Scholar 

  64. Grigg A, Begley CG, Juttner CA, et al. Effect of peripheral blood progenitor cells mobilised by filgrastim (G-CSF) on platelet recovery after high-dose chemotherapy. Bone Marrow Transplant 1993; 11 Suppl. 2: 23–9

    PubMed  Google Scholar 

  65. Sheridan WP, Begley CG, To LB, et al. Phase II study of autologous filgrastim (G-CSF)-mobilized peripheral blood progenitor cells to restore hemopoiesis after high-dose chemotherapy for lymphoid malignancies. Bone Marrow Transplant 1994; 14: 105–11

    CAS  PubMed  Google Scholar 

  66. Beyer J, Schwella N, Zingsem J, et al. Bone marrow versus peripheral blood stem cells as rescue after high-dose chemotherapy [abstract no. 1802]. Blood 1993; 82 Suppl. 1: 454a

    Google Scholar 

  67. Bolwell BJ, Fishleder A, Andresen SW, et al. G-CSF primed peripheral blood progenitor cells in autologous bone marrow transplantation: parameters affecting bone marrow engraftment. Bone Marrow Transplant 1993; 12: 609–14

    CAS  PubMed  Google Scholar 

  68. Hohaus S, Goldschmidt H, Erhardt R, et al. Successful autografting following myeloablative conditioning therapy with blood stem cells mobilized by chemotherapy plus rhG-CSF. Exp Hematol 1993; 21: 508–14

    CAS  PubMed  Google Scholar 

  69. Spitzer G, Spencer V, Dunphy F, et al. Are growth factors (GF) needed after peripheral blood stem cell (PBSC) transplantation? A randomised study to evaluate the question [abstract no. 1138]. Blood 1993; 82 Suppl. D: 288a

    Google Scholar 

  70. DeLuca E, Sheridan WP, Watson D, et al. Prior chemotherapy does not prevent effective mobilisation by G-CSF peripheral blood progenitor cells. Br J Cancer 1992; 66: 893–9

    CAS  PubMed  Google Scholar 

  71. Brugger W, Birken R, Bertz H, et al. Peripheral blood progenitor cells mobilized by chemotherapy plus granulocyte-colony stimulating factor accelerate both neutrophil and platelet recovery after high-dose VP16, ifosfamide and cisplatin. Br J Haematol 1993; 84: 402–7

    CAS  PubMed  Google Scholar 

  72. Pettengell R, Testa NG, Swindell R, et al. Transplantation potential of haematopoietic cells released into the circulation during routine chemotherapy for non-Hodgkin’s lymphoma. Blood 1993; 82: 2239–48

    CAS  PubMed  Google Scholar 

  73. Jones HM, Jones SA, Watts MJ, et al. Development of a simplified single-apheresis approach for peripheral-blood progenitor-cell transplantation in previously treated patients with lymphoma. J Clin Oncol 1994; 12: 1693–702

    CAS  PubMed  Google Scholar 

  74. Shimazaki C, Oku N, Ashihara E, et al. Collection of peripheral blood stem cells mobilized by high-dose Ara-C plus VP-16 or aclarubicin followed by recombinant human granulocyte-colony stimulating factor. Bone Marrow Transplant 1992; 10: 341–6

    CAS  PubMed  Google Scholar 

  75. Craig JIO, Anthony RS, Stewart A, et al. Peripheral blood stem cell mobilization using high-dose cyclophosphamide and G-CSF in pretreated patients with lymphoma. Br J Haematol 1993; 85: 210–2

    CAS  PubMed  Google Scholar 

  76. Dreger P, Marquardt P, Haferlach T, et al. Effective mobilisation of peripheral blood progenitor cells with ‘Dexa-BEAM’ and G-CSF: timing of harvesting and composition of the leukapheresis product. Br J Cancer 1993; 68: 950–7

    CAS  PubMed  Google Scholar 

  77. Demuynck H, Pettengell R, de Campos E, et al. The capacity of peripheral blood stem cells mobilised with chemotherapy plus G-CSF to repopulate irradiated marrow stroma in vitro is similar to that of bone marrow. Eur J Cancer 1992; 28: 381–6

    CAS  PubMed  Google Scholar 

  78. Fukuda M, Kojima S, Matsumoto K, et al. Autotransplantation of peripheral blood stem cells mobilized by chemotherapy and recombinant human granulocyte colony-stimulating factor in childhood neuroblastoma and non-Hodgkin’s lymphoma. Br J Haematol 1992; 80: 327–31

    CAS  PubMed  Google Scholar 

  79. Kawano Y, Takaue Y, Watanabe T, et al. Effects of progenitor cell dose and preleukapheresis use of recombinant granulocyte colony-stimulating factor on the recovery of hematopoiesis after blood stem cell autografting in children. Exp Hematol 1993; 21: 103–8

    CAS  PubMed  Google Scholar 

  80. Brugger W, Bross KJ, Glatt M, et al. Mobilization of tumor cells and hematopoietic progenitor cells into peripheral blood of patients with solid tumors. Blood 1994; 83: 636–40

    CAS  PubMed  Google Scholar 

  81. Vora AJ, Toh CH, Peel J, et al. Use of granulocyte colony-stimulating factor (G-CSF) for mobilizing peripheral blood stem cells: risk of mobilizing clonal myeloma cells in patients with bone marrow infiltration. Br J Haematol 1994; 86: 180–2

    CAS  PubMed  Google Scholar 

  82. Hénon PR. Peripheral blood stem cell transplantations: past, present and future. Stem Cells 1993; 11: 154–72

    PubMed  Google Scholar 

  83. Negrin RS, Haeuber DH, Nagler A, et al. Treatment of myelodysplastic syndromes with recombinant human granulocyte colony-stimulating factor: a phase I-II trial. Ann Intern Med 1989; 110: 976–84

    CAS  PubMed  Google Scholar 

  84. Greenberg P, Taylor K, Larson R, et al. Phase III randomised multicentre trial of G-CSF vs observation for myelodysplastic syndromes (MDS) [abstract]. Blood 1993; 82 Suppl. 1: 768

    Google Scholar 

  85. Negrin RS, Stein R, Vardiman J, et al. Treatment of the anaemia of myelodysplastic syndromes using recombinant human granulocyte colony-stimulating factor in combination with erythropoietin. Blood 1993; 82: 737–43

    CAS  PubMed  Google Scholar 

  86. Schriber JR, Negrin RS. Use and toxicity of the colony-stimulating factors. Drug Saf 1993; 8: 457–68

    CAS  PubMed  Google Scholar 

  87. Scherrer R, Geissler K, Kyrle PA, et al. Granulocyte colony-stimulating factor (G-CSF) as an adjunct to induction chemotherapy of adult acute lymphoblastic leukemia (ALL). Ann Hematol 1993; 66: 283–9

    CAS  PubMed  Google Scholar 

  88. Ohno R, Tomonaga M, Kobayashi T, et al. Effect of granulocyte colony-stimulating factor after intensive induction chemotherapy in relapsed or refractory acute leukemia. N Engl J Med 1990; 323: 871–7

    CAS  PubMed  Google Scholar 

  89. Glaspy JA, Baldwin GC, Robertson PA, et al. Therapy for neutropenia in hairy cell leukemia with recombinant human granulocyte colony-stimulating factor. Ann Intern Med 1988; 109: 789–95

    CAS  PubMed  Google Scholar 

  90. Glaspy JA, Souza L, Scates S, et al. Treatment of hairy cell leukemia with granulocyte colony-stimulating factor and recombinantinterferon-alpha-2b. J Immunol 1992; 11: 198–208

    CAS  Google Scholar 

  91. Dale DC, Hammond WP, Gabrilove J, et al. Long term treatment of severe chronic neutropenia with recombinant human granulocyte colony-stimulating factor (rmetHuG-CSF) [abstract]. Blood 1990; 76: 139a

    Google Scholar 

  92. Dale DC, Bonilla MA, Davis MW, et al. A randomized controlled phase III trial of recombinant human granulocyte colony-stimulating factor (filgrastim) for treatment of severe chronic neutropenia. Blood 1993; 81: 2496–502

    CAS  PubMed  Google Scholar 

  93. Boxer LA, Hutchinson R, Emerson S. Recombinant human granulocyte-colony-stimulating factor in the treatment of patients with neutropenia. Clin Immunol Immunopathol 1992; 62: S39–46

    CAS  PubMed  Google Scholar 

  94. Besner GE, Glick PL, Karp MP, et al. Recombinant human granulocyte colony-stimulating factor promotes wound healing in a patient with congenital neutropenia. J Pediatr Surg 1992; 27: 288–91

    CAS  PubMed  Google Scholar 

  95. Fasth A, Carlson G, Ridell B. Rekombinant granulocyt-kolonistimulerande faktor ny effektiv behandling av grav neutropeni. Lakartidningen 1992; 89: 776–80

    CAS  PubMed  Google Scholar 

  96. Freund M, Heussner P, Kanz L, et al. Granulocyte colony-stimulating factor (G-CSF) for the treatment of adult patients with chronic idiopathic neutropenia and cyclic neutropenia [abstract no. 122]. Onkologie 1991; 14 Suppl. 2: 43–4

    Google Scholar 

  97. Furukawa T, Takahashi M, Moriyama Y, et al. Successful treatment of chronic idiopathic neutropenia using recombinant granulocyte colony-stimulating factor. Ann Hematol 1991; 62: 22–4

    CAS  PubMed  Google Scholar 

  98. Imashuku S, Tsuchida M, Sasaki M, et al. Recombinant human granulocyte colony-stimulating factor in the treatment of patients with chronic benign granulocytopenia and congenital agranulocytosis (Kostmann’s syndrome). Acta Paediatr 1992; 81: 133–6

    CAS  PubMed  Google Scholar 

  99. Marlton PV, Wright SJ, Taylor KM. Granulocyte colony stimulating factor in the management of chronic neutropenia. Med J Aust 1992; 156: 729–31

    CAS  PubMed  Google Scholar 

  100. Weston B, Axtell RA, Todd III RF, et al. Clinical and biologic effects of granulocyte colony-stimulating factor in the treatment of myelokathexis. J Pediatr 1991; 118: 229–34

    CAS  PubMed  Google Scholar 

  101. Zeidler C, Reiter A, Yakisan E, et al. Effects of r-met HuG-CSF treatment in patients with severe congenital neutropenia [in German]. Klin Padiatr 1993; 205: 264–71

    CAS  PubMed  Google Scholar 

  102. Jones EA, Bolyard AA, Dale DC. Quality of life of patients with severe chronic neutropenia receiving long-term treatment with granulocyte colony-stimulating factor. JAMA 1993; 270: 1132–3

    CAS  PubMed  Google Scholar 

  103. Fazio MT, Glaspy JA. The impact of granulocyte colony-stimulating factor on quality of life in patients with severe chronic neutropenia. Oncol Nurse Forum 1991; 18: 1411–4

    CAS  Google Scholar 

  104. Israel DS, Plaisance Kl. Neutropenia in patients infected with human immunodeficiency virus. Clin Pharm 1991; 10: 268–79

    CAS  PubMed  Google Scholar 

  105. Mitsuyasu RT, Miles SA, Golde DW. The use of myeloid hematopoietic growth factors in patients with HIV infection. Int J Cell Cloning 1990; 8 Suppl. 1: 347–55

    PubMed  Google Scholar 

  106. Miles SA. The use of hematopoietic growth factors in HIV infection and AIDS-related malignancies. Cancer Invest 1991; 9: 229–38

    CAS  PubMed  Google Scholar 

  107. Jacobson MA, Stanley HD, Heard SE. Ganiciclovir with recombinant methionyl human granulocyte colony-stimulating factor for treatment of cytomegalovirus disease in AIDS patients [letter]. AIDS 1992; 6: 515–7

    CAS  PubMed  Google Scholar 

  108. Miles SA, Mitsuyasu RT, Moreno J, et al. Combined therapy with recombinant granulocyte colony-stimulating factor and erythropoietin decreases hematologic toxicity from zidovudine. Blood 1991; 77: 2109–77

    CAS  PubMed  Google Scholar 

  109. Levi GC, Katz A. The role of G-CSF in zidovudine-induced leukopenia and granulocytopenia in AIDS patients: results of a pilot study. Int J Immunother 1992; 8: 203–6

    Google Scholar 

  110. Mueller BU, Jacobsen F, Butler KM, et al. Combination treatment with azidothymidine and granulocyte colony-stimulating factor in children with human immunodeficiency virus infection. J Pediatr 1992; 121: 797–802

    CAS  PubMed  Google Scholar 

  111. Hermans P, Franchioly P, Thioux C, et al. Granulocyte-colony stimulating factor (G-CSF [filgrastim]) for treatment of neutropenia in patients with AIDS [abstract no. WS-B22-3]. In: IXth International Conference on AIDS in affiliation with the IVth STD World Congress: June 6–11, 1993. Berlin:, 1993: 65

  112. Zuccotti GV, Flumine P, Locatelli V, et al. Growth factors and HIV-infection in children. J Int Med Res 1993; 21: 342–5

    CAS  PubMed  Google Scholar 

  113. Hengge UR, Brockmeyer NH, Goos M. Granulocyte colony-stimulating factor treatment in AIDS patients. Clin Investig 1992; 70: 922–6

    CAS  PubMed  Google Scholar 

  114. Geibig CB, Marks LW. Treatment of clozapine- and molindone-induced agranulocytosis with granulocyte colony-stimulating factor. Ann Pharmacother 1993; 27: 1190–2

    CAS  PubMed  Google Scholar 

  115. Gerson SL, Gullion G, Yeh H-S, et al. Granulocyte colony-stimulating factor for clozapine-induced agranulocytosis [letter]. Lancet 1992; 340: 1097

    CAS  PubMed  Google Scholar 

  116. Nielsen H. Recombinant human granulocyte colony-stimulating factor (rhG-CSF; filgrastim) treatment of clozapine-induced agranulocytosis. J Intern Med 1993; 234: 529–31

    CAS  PubMed  Google Scholar 

  117. Chia HM, Kalra L, Lakhani AK, et al. Filgrastim for low-dose, captopril-induced agranulocytosis [letter]. Lancet 1993; 342: 304

    CAS  PubMed  Google Scholar 

  118. Collins DA, Tobias JH, Hill RP, et al. Reversal of gold-induced neutropenia with granulocyte colony-stimulating factor (G-CSF). Br J Rheumatol 1993; 32: 518–20

    CAS  PubMed  Google Scholar 

  119. Teitelbaum AH, Bell AJ, Brown SL. Filgrastim (r-metHuG-CSF) reversal of drug-induced agranulocytosis [letter]. Am J Med 1993; 95: 245–6

    CAS  PubMed  Google Scholar 

  120. Balkin MS, Buchholtz M, Ortiz J, et al. Propythiouracil (PTU)-induced agranulocytosis treated with recombinant human granulocyte colony-stimulating factor (G-CSF). Thyroid 1993; 3: 305–9

    CAS  PubMed  Google Scholar 

  121. Wyatt S, Joyner MV, Daneshmend TK. Filgrastim for mesalazine-associated neutropenia [letter]. Lancet 1993; 341: 1476

    CAS  PubMed  Google Scholar 

  122. Marks LB, Friedman HS, Kurtzberg J, et al. Reversal of radiation-induced neutropenia by granulocyte colony-stimulating factor. Med Pediatr Oncol 1992; 20: 240–2

    CAS  PubMed  Google Scholar 

  123. MacManus MP, Clarke J, McCormick D, et al. Use of recombinant granulocyte colony-stimulating factor to treat neutropenia occurring during craniospinal irradiation. Int J Radiat Oncol Biol Phys 1993; 26: 845–50

    CAS  Google Scholar 

  124. Bamberg M, Schmidberger H, Hess CF. G-CSF in the management of neutropenia during radiotherapy. Tumordiagn Ther 1992; 13: 125–6

    Google Scholar 

  125. Colquhoun SD, Snaked A, Jurim O, et al. Reversal of neutropenia with granulocyte colony-stimulating factor without precipitating liver allograft rejection [letter]. Transplantation 1993; 56: 755–7

    Google Scholar 

  126. Gordon MS, O’Donnell JA, Mohler III ER, et al. The use of granulocyte colony-stimulating factor in the treatment of fever and neutropenia in a heart transplant patient. J Heart Lung Transplant 1993; 12: 706–7

    CAS  PubMed  Google Scholar 

  127. Roberts RL, Szelc CM, Scates SM, et al. Neutropenia in an extremely premature infant treated with recombinant human granulocyte colony-stimulating factor. Am J Dis Child 1991; 145: 808–12

    CAS  PubMed  Google Scholar 

  128. Wang WC, Cordoba J, Infante AJ, et al. Successful treatment of neutropenia in the hyper-immunoglobulin M syndrome with granulocyte colony-stimulating factor. Am J Pediatr Hematol Oncol 1994; 16: 160–3

    CAS  PubMed  Google Scholar 

  129. van de Merwe JP, Hooijkaas H. Sjögren’s syndrome in patients with chronic bacterial respiratory infections and a diminished chemotactic responsiveness of monocytes and/or granulocytes: treatment with filgrastim (G-CSF) [abstract no. 53]. Clin Rheumatol 1994; 13: 161

    Google Scholar 

  130. Wagner DR, Combe C, Gresser U. GM-CSF and G-CSF in Felty’s syndrome. Clin Investig 1993; 71: 168–71

    CAS  PubMed  Google Scholar 

  131. Lang DF, Rosenfeld CS, Diamond HS, et al. Successful treatment of Tγ lymphoproliferative disease with human-recombinant granulocyte colony-stimulating factor. Am J Hematol 1992; 40: 66–8

    CAS  PubMed  Google Scholar 

  132. Decoster G, Rich W, Brown S. Safety profile of filgrastim. In: Morstyn G, Dexter M, editors. Filgrastim in clinical practice. New York: Marcel Dekker, 1993: 267–90

    Google Scholar 

  133. Oeda E, Shinohara K, Kamei S, et al. Capillary leak syndrome likely the result of granulocyte colony-stimulating factor after high-dose chemotherapy. Intern Med 1994; 33: 115–9

    CAS  PubMed  Google Scholar 

  134. Schliesser G, Pralle H, Lohmeyer J. Leukocytoclastic vasculitis complicating granulocyte colony-stimulating factor (G-CSF) induced neutrophil recovery in Ty-lymphocytosis with severe neutropenia. Ann Hematol 1992; 65: 151–2

    CAS  PubMed  Google Scholar 

  135. Jaiyesimi I, Giralt SS, Wood J. Subcutaneous granulocyte colony-stimulating factor and acute anaphylaxis [letter]. N Engl J Med 1991; 325: 587

    CAS  PubMed  Google Scholar 

  136. Brown SL, Hill E. Subcutaneous granulocyte colony-stimulating factor and acute anaphylaxis. Reply [letter]. N Engl J Med 1991; 325: 587

    Google Scholar 

  137. Rivera J, Zuazu I, Sánchez MJ, et al. Effect of G-CSF on the haematopoietic system [letter]. Thromb Haemost 1994; 71: 804–5

    CAS  PubMed  Google Scholar 

  138. Kawachi Y, Ozaki S, Sakamoto Y, et al. Richter’s syndrome showing pronounced lymphadenopathy in response to administration of granulocyte colony-stimulating factor. Leuk Lymphoma 1994; 13: 509–14

    CAS  PubMed  Google Scholar 

  139. Filgrastim prescribing information. Kirin-Sankyo, Japan, October 1992.

  140. Livingston RB. Dose-intensity and high dose therapy: two different concepts. Cancer 1994; 74: 1177–83

    CAS  PubMed  Google Scholar 

  141. Triozzi PL. Autologous bone marrow transplantation and peripheral blood progenitor transplant for breast cancer. Lancet 1994; 344: 418–9

    CAS  PubMed  Google Scholar 

  142. Leyvraz S, Ketterer N, Vuichard P, et al. Sequential high-dose combination chemotherapy with granulocyte colony-stimulating factor and peripheral blood progenitor cells in patients with solid tumours: intensification limited by nonhematologic effects [letter]. J Natl Cancer Inst 1993; 85: 1962–4

    CAS  PubMed  Google Scholar 

  143. Nissen C, Dalle Carbonare V, Moser Y. In vitro comparison of the biological potency of glycosylated versus non-glycosylated rG-CSF. Drug Invest 1994; 7: 346–52

    CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Additional information

Various sections of the manuscript reviewed by: W.M. Bennett, Division of Nephrology, Hypertension and Clinical Pharmacology, Oregon Health Sciences University, Portland, Oregon, USA; L.A. Boxer, Department of Pediatrics, C.S. Mott Children’s Hospital, Ann Arbor, Michigan, USA; D.C. Dale, Department of Medicine, University of Washington, Seattle, Washington, USA; D. Dunlop, CRC Department of Medical Oncology, University of Glasgow, Glasgow, Scotland; K. Eguchi, Department of Internal Medicine and Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; W.E. Evans, Pharmaceutical Department, St Jude Children’s Research Hospital, Memphis, Tennessee, USA; R. Haas, Universität Heidelberg, Heidelberg, Germany; Y. Itoh, The First Department of Internal Medicine, Tokyo Medical College, Tokyo, Japan; D.C. Lynch, Department of Haematology, University College London Medical School, London, England; E. Masson, Pharmaceutical Department, St Jude Children’s Research Hospital, Memphis, Tennessee, USA; R. Pettengell, Developmental Hematopoiesis Laboratory, Sloan-Kettering Institute for Cancer Research, New York, New York, USA.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Frampton, J.E., Lee, C.R. & Faulds, D. Filgrastim. Drugs 48, 731–760 (1994). https://doi.org/10.2165/00003495-199448050-00007

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-199448050-00007

Keywords

Navigation