Take a look at the Recent articles

Immunosuppressive agents in hematopoietic stem cell transplantation

Eren Arslan Davulcu

Ege University Faculty of Medicine, Hematology Department, Bornova, İzmir, Turkey

E-mail : aa

Filiz Vural

Ege University Faculty of Medicine, Hematology Department, Bornova, İzmir, Turkey

DOI: 10.15761/TiT.1000240

Article
Article Info
Author Info
Figures & Data

Abstract

Hematopoietic stem cell transplantation has been used in many malignant and non-malignant diseases successfully. Allogeneic stem cell transplantation has its own immunologic characteristics such as graft versus host disease. In order to prevent graft versus host disease, immunosuppressive medications are required. Immunosuppressive drugs have diverse mechanisms of action thus they could be used in different clinical status. Steroids and methotrexate are nonspecific; cyclosporine, tacrolimus, sirolimus and mycophenolate are T cell specific immunosuppressive drugs. Antibodies, monoclonal antibodies, and new drugs such as ruxolitinib are available treatment options but we still need improvement and innovation in management of graft versus host disease.

Key words

allogeneic stem cell transplantation, graft versus host disease, hematopoietic stem cell transplantation, immunosuppressive agents

Introduction

Hematopoietic stem cell transplantation (HSCT) is a curative treatment modality for a wide range of malignant and non-malignant diseases. Immunosuppressive treatment is required following allogeneic hematopoietic stem cell transplantation (allo-HSCT) in order to prevent graft versus host disease (GVHD) and associated organ-tissue lost. Donor T cells recognize different histocompatibility antigens of the host cells as foreign. As a result of antigen recognition, activated Tcells secrete cytokines such as interleukin (IL) -1, IL-2, tumor necrosis factor (TNF), and gamma-interferon, so that GVHD process begin [1]. GVHD is classified as acute and chronic according to time of onset. Classically acute GVHD occurred within 100 days after transplantation. Rarely, patients may present acute GVHD findings after than 100 days which is named 'persistent, recurrent, late onset acute GVHD'. 'Overlap syndrome' is another clinical scenario which may carry both acute and chronic GVHD features at any time after HSCT [2]. Numerous risk factors have been identified for development of acute GVHD, one of the most important one is acute GVHD prophylactic regimen used [3]. Immunosuppressive agents for prophylaxis and treatment of GVHD are evaluated in two major categories: 1- Non-specific immunosupressives, 2- T cell mediated drugs, 3- Antibodies, 4- Other drugs.

Non-specific immunosuppressive drugs

a-Corticosteroids:

Although mechanism of action is not fully understood, corticosteroids are agents that are preferentially used and combined with other immunosuppressive in acute GVHD treatment. Corticosteroids are thought to reduce T cell numbers, diminish expression and production of inflammatory cytokines such as IL-1 and TNF alfa (1), and inhibits cytokine gene transcription [4].

In acute GVHD treatment, intravenous methylprednisolon is preferred because patients may not be able to take oral medication or there may be malabsorbtion because of severe mucositis. If oral medication is available oral methylprednisolone or rarely oral prednisone can be prescribed.

The classical, first-line treatment in acute GVHD is methylprednisolone. It should be initiated if GVHD is grade 2 or higher and initilal dose is 2 mg/kg/day given in two divided doses. Initial dose is recommended to be continued for 7 days. Tapering strategy is based on the clinical response and it should not be stopped until all signs of GVHD are disappeared. In gastrointestinal GVHD, oral budesonid may be given synchronous with methylprednisolone [5].

Methylprednisolon is preferred in GVDH prophylaxis in combination with other immunosuppressive drugs. Adding steroid provides reducing immunosuppressant-related toxicity without altering the efficacy [6].

Corticosteroids have several side effects such as making susceptibility to infections, Cushingoid appearance, psychosis, hyperglycemia, hypertension, osteoporosis, bone aseptic necrosis, and cataract formation.

b-Methotrexate:

Methotrexate is a folic acid antagonist and inhibits dihydrofolate reductase (DHFR) which converts folic acid to reduced tetrahydrofolate. Tetrahydrofolate transfers single carbon groups which are used for purine and thymidylate synthesis [7]. Eventually, methotrexate prevents nucleotide synthesis. Lymphocytes are relatively more prone to methotrexate because intracellular polyglutamate formation is more prominent in lymphocytes which is toxic for DHFR and augments antifolate activity [8]. In GVHD prophylaxismethotrexate is thought to reduce activated T cell proliferation.

The most common side effects are renal, gastrointestinal, and hepatic toxicity. In patients with renal failure, methotrexate may be more toxic and dose reduction is necessary. If a there is a fluid collection (ascites, pericardial or pleural effusion) in any part of the body, methotrexate should not be administered because the increased risk of acute renal failure.

Methotrexate classically administered intravenously on days +1, +3, +6 and +11after allo-HSCT, in combination with cyclosporine [9].

Specific T cell immunosuppressive drugs

a-Cyclosporine:

It is a calcineurin inhibitor that is extracted from fungi, blocks the transcription of cytokine genes in activated T cells. Cyclosporin makes a complex with cyclophilin, inhibits the phosphatase activity of calcineurin which regulates nuclear translocation and activation of NFAT (nuclear factor of activate T cells) transcription factors. Furthermore, cyclosporine blocks the activation of JNK (Jun N-terminal kinase) and p38 signaling pathways triggered by antigen recognition, making cyclosporin a highly specific inhibitor of T cell activation [10]. The analogous cytosolic protein for the drug tacrolimus is FK binding protein (FKBP). Both cyclophilin and FKBP function as the enzyme prolylcis-trans isomerase (another name rotamase) [11]. Rotamase takes part in protein folding process. Inhibition of rotamase by cyclosporin and tacrolimus is thought to be important step in immunosuppression.

Major side effects are nephro and hepatotoxicity, hypertension, hyperglycemia, headaches, hirsutism, gum hypertrophy, brittle nails, acne, nausea, vomiting, neurotoxicity, and thrombotic microangiopathy. Nephrotoxicity is dose related and associated with vasoconstriction and ischemia of the afferent arterioles in the kidney [12,13]. It should be kept in mind that there may be interactions between cyclosporine and drugs metabolized by cytochrome p450 enzymes such as voriconazole.

In myeloablative conditioning regimens, the standard GVHD prophylaxis is cyclosporine and short course of methotrexate. The initial cyclosporine dose is 3 mg/kg/day starting on the preceding the infusion of the stem cells (day -1). It is given as short intravenous bolus, preferably in two divided doses. If oral intake is available, we can change the route by giving the first oral dose twice of the last intravenous dose. Dose adjustment is necessary according to blood drug concentration and toxicities. The target cyclosporine concentration is 200–300 mg/L during the first 3–4 weeks, then 100–200 mg/L until 3 months after transplantation if there is no GVHD or toxicity. Drug concentration should be measured after 12 hours from administration. Prophylaxis is continued until 6 months if GVHD is absent. Tapering the dose is possible after 3 months in the absence of GVHD [5]. Cyclosporine prophylaxis regimen is the same in reduced intensity conditioning allo-HSCTs.

b- Tacrolimus (FK506):

Mechanism of action, administration, and clinical use are quite similar to cyclosporine. Major side effects are neuro and nephrotoxicity, hyperglycemia, dyspnea, musculoskeletal pain, anorexia, nausea, vomiting, abdominal pain, itching, fatigue, and thrombotic microangiopathy. Central nervous system effects are in wide spectrum such as headaches, tremors, paresthesias, seizures, photophobia, mental status changes, mutism, and encephalopathy [14].

Cyclosporine and tacrolimus are accepted as equivalent in GVHD prophylaxis, but the latter is less frequently used and experienced in Europe [5].

Its dose in GVHD prophylaxis is 0.03 to 0.04 mg/kg per day as a continuous infusion. When oral intake is available 0.15 mg/kg per day administered in two divided doses. The target blood concentration is 10 -30 ng/mL.

c-Sirolimus:

Sirolimus (rapamycin) is a macrolide that is currently approved by the FDA only in solid organ transplantation in order to prevent organ rejection.

d-Mycophenolate:

There are two forms available: 1-Mycophenolate mofetil (MMF): Morpholinoethyl ester of mycophenolic acid (MPA), 2- Sodium salt of mycophenolate. MMF is a prodrug of MPA, which is an inhibitor of inosine monophosphate dehydrogenase. This enzyme is rate-limiting in de novo synthesis of guanosine nucleotides. T and B lymphocytes are more dependent on de novo pathway than other cells [15].

Major side effects are neutropenia, hyper or hypotension, abdominal pain, hyperglycemia, and nephrotoxicity.

The standard GVHD prophylaxis in reduced intensity conditioning regimen is cyclosporine and MMF combination. The standard dose is 30 mg/kg/day, orally, in two divided doses. Administration is started on day +1 and continued. Dose adjustment according to toxicity is required. Treatment should be continued 1 month in sibling transplantations, 3 months in unrelated or mismatched donor transplantations [5].

Antibodies

a- Antithymocyte Globulin (ATG):

It is a polyclonal immune globulin can be produced by injection of thymocytes to horses or rabbits, and by using Jurkat cells as an antigen. Because of the polyclonal nature of ATG, has diverse effects on the immune system. ATG makes complement mediated T-cell depletion, modulates surface molecules of the leukocytes and endothelial cells, stimulates B cell apoptosis, changes dendritic cell functions, and induces regulatory T cells and natural killer cells [16].

ATG is recommended in GVHD prophylaxis of unrelated donor transplantations either in myeloablative or non-myeloablative regimens. Doses of two different brands are: ATG-Frensius is 10 mg/kg for 3 days (total dose 30 mg/kg) and Thymoglobulin is 2.5 mg/kg for 3 days (total dose 7.5 mg/kg), both administered on days -3, -2, and -1. ATG may also be used in treatment of acute GVHD as a second-line treatment [5].

Other drugs

a-Rituximab:

Adding anti CD-20 monoclonal antibody rituximab into preparative regimens or chronic GVHD treatment protocols is thought to suppress donor B cell immunity. At present, rituximab is not standard drug in HSCT era. It may be an option in second-line chronic GVHD treatment.

b-Ruxolitinib:

A selective Janus kinase (JAK) 1/2 inhibitor ruxolitinib is an oral drug used in myelofibrosis treatment, have recently been discovered to be beneficial in acute and chronic GVHD treatment. JAK 1/2 takes place in multiple steps in GVHD related inflammation and tissue damage such as T cell activation, lineage formation and survival through the common gamma chain of six different interleukins (IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21) [17], activation of neutrophils [18], and dendritic cells [19].

A recent, large multicenter study pointed that ruxolitinib has high response and 6-month survival rates in steroid refractory acute and chronic GVHD patients [20].

Anti tumor necrosis factor agents, extracorporeal photopheresis, and mesenchymal stem cell infusion are some of the alternative treatment modalities for chronic GVHD [5].

There is no standard approach in chronic GVHD treatment, therefore transplantation centers should choose the appropriate one according to their clinical practice, availability of drugs, and institutional guidelines.

References

  1. Antin JH, Ferrara JL (1992) Cytokine dysregulation and acute graft-versus-host disease. Blood 80: 2964-2968. [Crossref]
  2. Filipovich AH, Weisdorf D, Pavletic S, Socie G, Wingard JR, et al. (2005) National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report. Biol Blood Marrow Transplant 11(12): 945.
  3. Gale RP, Bortin MM, van Bekkum DW, Biggs JC, Dicke KA, et al. (1987) Risk factors for acute graft-versus-host disease. Br J Haematol 67: 397-406. [Crossref]
  4. Almawi WY, Lipman ML, Stevens AC, Zanker B, Hadro ET, et al. (1991) Abrogation of glucocorticoid-mediated inhibition of T cell proliferation by the synergistic action of IL-1, IL-6, and IFN-gamma. J Immunol 146(10): 3523.
  5. Ruutu T, Gratwohl A, de Witte T, Afanasyev B, Apperley J, et al. (2014) Prophylaxis and treatment of GVHD: EBMT-ELN working group recommendations for a standardized practice. Bone Marrow Transplant 49(2):168-73.
  6. Yau JC, Dimopoulos MA, Huan SD, Spencer V, Woo SY, et al. (1991) An effective acute graft-vs.-host disease prophylaxis with mini dose methotrexate, cyclosporine, and single-dose methylprednisolone. Am J Hematol 38(4): 288-292.
  7. Zaharko DS, Fung WP, Yang KH (1977) Relative biochemical aspects of low and high doses of methotrexate in mice. Cancer Res 37: 1602-1607. [Crossref]
  8. Fabre G, Goldman ID (1985) Formation of 7-hydroxymethotrexate polyglutamyl derivatives and their cytotoxicity in human chronic myelogenous leukemia cells, in vitro. Cancer Res 45(1):80.
  9. Storb R, Deeg HJ, Whitehead J, Appelbaum F, Beatty P, et al. (1986) Methotrexate and cyclosporine compared with cyclosporine alone for prophylaxis of acute graft versus host disease after marrow transplantation for leukemia. N Engl J Med 314(12): 729.
  10. Matsuda S, Koyasu S (2000) Mechanisms of action of cyclosporine. Immunopharmacology 47: 119-125. [Crossref]
  11. Gething MJ, Sambrook J (1992) Protein folding in the cell. Nature 355: 33-45. [Crossref]
  12. Bennett WM, Pulliam JP (1983) Cyclosporine nephrotoxicity. Ann Intern Med 99: 851-854. [Crossref]
  13. Petric R, Freeman D, Wallace C, McDonald J, Stiller C, et al. 1988. Effect of cyclosporine on urinary prostanoid excretion, renal blood flow, and glomerulotubular function. Transplantation 45(5): 883.
  14. Eidelman BH, Abu-Elmagd K, Wilson J, Fung JJ, Alessiani M, et al. (1991) Neurologic complications of FK 506. Transplant Proc 23: 3175-3178. [Crossref]
  15. Allison AC, Eugui EM (2000) Mycophenolate mofetil and its mechanisms of action. Immunopharmacology 47: 85-118. [Crossref]
  16. Mohty M (2007) Mechanisms of action of antithymocyte globulin: T-cell depletion and beyond. Leukemia 21: 1387-1394. [Crossref]
  17. He YW, Adkins B, Furse RK, Malek TR (1995) Expression and function of the gamma c subunit of the IL-2, IL-4, and IL-7 receptors. Distinct interaction of gamma c in the IL-4 receptor. J Immunol 154: 1596-1606.
  18. Nicholson SE, Oates AC, Harpur AG, Ziemiecki A, Wilks AF, et al. (1994) Tyrosine kinase JAK1 is associated with the granulocyte-colony-stimulating factor receptor and both become tyrosine-phosphorylated after receptor activation. Proc Natl Acad Sci U S A 91: 2985-2988.
  19. Heine A, Held SA, Daecke SN, Wallner S, Yajnanarayana SP, et al. (2013) The JAK-inhibitor ruxolitinib impairs dendritic cell function in vitro and in vivo. Blood 122: 1192-1202. [Crossref]
  20. Zeiser R, Burchert A, Lengerke C, Verbeek M, Maas-Bauer K, et al. (2015) Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey. Leukemia 29(10): 2062-2068.

Editorial Information

Editor-in-Chief

Transplantation Open

Article Type

Short Communication

Publication History

Received: October 27, 2017
Accepted: November 24, 2017
Published: November 27, 2017

Copyright

©2017 Davulcu EA. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Davulcu EA, Vural F (2017) Immunosuppressive agents in hematopoietic stem cell transplantation. Trends in Transplant 11. DOI: 10.15761/TiT.1000240

Corresponding author

Filiz Vural

Ege University Hematology Department, Bornova, İzmir, Turkey, Tel: +90 23238904437, +90 2323738797

No Data.