Skip to main content
American Journal of Blood Research logoLink to American Journal of Blood Research
. 2011 Jun 1;1(1):57–64.

The evolving role of statins in hematopoietic stem and progenitor cell transplantation

Mehdi Hamadani 1, Michael D Craig 1, Laura F Gibson 1, Scot C Remick 1
PMCID: PMC3301410  PMID: 22432066

Abstract

Allogeneic hematopoietic stem cell transplantation is the sole curative modality for a variety of malignant and benign hematological disorders. Despite advances in supportive care and transplant conditioning regimens graft-versus-host disease (GVHD), infectious complications and end organ toxicity remain the leading causes of transplant related mortality (TRM). Development of safe and effective strategies to mitigate these significant complications associated with HSCT, are urgently needed. Statins are lipid lowering drugs, which reduce cholesterol production by inhibiting HMG-CoA reductase, with a well defined toxicity profile. Statins have pleiotropic immunomodulatory effects which are relevant in the context of treating and preventing GVHD. In addition to GVHD statins may possess several other effects that might have clinical benefit in the setting of hematopoietic cell transplantation, such as treatment of bronchiolitis obliterans and antineoplastic activity. Herein we review the emerging role of statins in improving the outcomes of patients undergoing HSCT.

Keywords: Statin, atorvastatin, GVHD, HMG-CoA reductase inhibitors, allogeneic stem cell transplantation, multiple myeloma, idiopathic pulmonary fibrosis, graft-versus-host disease

Introduction

Allogeneic hematopoietic stem cell transplantation (HSCT) is the sole curative modality for a variety of malignant and benign hematological disorders [1]. Despite advances in supportive care and transplant conditioning regimens graft-versus-host disease (GVHD), infectious complications and end organ toxicity remain the leading causes of transplant related mortality (TRM) [2]. Development of safe and effective strategies to mitigate these significant complications associated with HSCT, are urgently needed. Herein we review the emerging role of statins in improving the outcomes of patients undergoing HSCT.

HMG-CoA reductase inhibitors

HMG-CoA (3-Hydroxy-3-MethylGlutaryl-Coenzyme-A) reductase inhibitors (statins) are prescribed worldwide to prevent and treat dyslipidemia and atherosclerotic vascular disease [3]. The mechanism of action involves binding to HMG-CoA reductase and displacement of its natural substrate HMG-CoA, leading to inhibition of mevalonate and cholesterol biosynthesis [3]. Atorvastatin, the prototypic statin drug has a proven safety track record [4-6]. Comprehensive assessment of atorvastatin's safety profile using data from 44 clinical trials comprising 16,495 patients demonstrated that the incidence of adverse events with atorvastatin did not increase in the 10 to 80 mg dose range and was similar to that observed with placebo. Moreover musculoskeletal and hepatic adverse events were infrequent and rarely required treatment discontinuation [7].

Graft-versus-Host Disease

GVHD develops in 30-75% of transplant recipients depending on the degree of histocompati-bility between the donor and the recipient, donor's age, ABO compatibility, GVHD prophylaxis used etc. [8, 9]. With standard (calcineurin inhibitor ± methotrexate or mycophenolate mofetil-based) prophylactic regimens, GVHD continues to be a major transplant complication. More intense (and often expensive) GVHD prophylactic strategies employing ex vivo T-cell depletion (TCD), and monoclonal or polyclonal antibodies, often display a narrow therapeutic index and are frequently complicated by delayed immune-reconstitution, increased risk of infectious complications and disease relapse [10-13], adding significant burden [14] to already enormous healthcare costs of transplant recipients [15, 16]. To date GVHD remains the major barrier to successful allogeneic HSCT outcomes, underscoring the urgent need for novel strategies designed to effectively prevent the development of this life threatening complication.

Immunomodulatory effects of Statins and GVHD

Statins possess potent immunomodulatory and anti-inflammatory properties that are critical not only in atherosclerosis-associated inflammation but are also relevant in autoimmune diseases such as multiple sclerosis and rheumatoid arthritis [17-19]. Reduced mevalonate production by statins not only inhibits the synthesis of cholesterol, but it also inhibits production of key isoprenoid intermediate molecules required for the isoprenylation of GTP-binding cell signaling proteins such as Ras, Rho and Rac [3]. By inhibiting this posttranslational modification, statins affect multiple intracellular signaling pathways and cellular functions including differentiation, motility, secretion and proliferation [19, 20].

Statin-mediated inhibition of Ras may lead to a bias towards T-helper type 2 (Th-2) cell development and inhibition of pro-inflammatory Th-1 driven responses [21]. Acute GVHD is thought to be mediated by alloreactive T cells with a Th-1 cytokine phenotype. Interestingly polarized Th-2 alloreactive CD4+ donor T cells fail to induce experimental acute GVHD [22-24]. Statin mediated changes in intracellular signaling can also affect the development of regulatory T (Treg) cells, as exposure of human CD4+ T cells to atorvastatin in vitro increases expression of forkhead box P3 (FOXP3) [25]. In fact increased numbers of circulating FOXP3+ Treg cells are present in dyslipidemic patients taking statins [25]. Statins also promote the migration of Treg cells to inflammatory foci in skin by increased expression of CXCL1, the ligand for CCR8, a chemokine receptor that is highly expressed on FOXP3+ Treg cells [26]. Expansion of Treg cells is an attractive strategy to prevent acute GVHD [27].

Statins indirectly decrease T-cell activation by inhibiting interferon gamma (IFN-γ) induced expression of major histocompatibility complex class (MHC) II molecules and by blocking up-regulation of a variety of co-stimulatory molecules and cytokines in antigen presenting cells (APCs) [19, 28]. APCs are critical for the pathogenesis of GVHD, and strategies to prevent activation of APCs can potentially abrogate GVHD [29]. In summary, statins demonstrate the ability to suppress T cell-dependent immune responses at multiple levels and hold great promise as a new class of immunomodulatory drugs to prevent acute GVHD.

Statins and experimental acute GVHD

Statins (atorvastatin and fluvastatin) have shown the ability to prevent acute GVHD in a MHC mismatched murine bone marrow transplant (BMT) model [30]. In a series of elegant experiments, Zeiser et al. demonstrated that pre-treating donor mice with atorvastatin lead to significantly less acute GVHD by: (i) inhibiting donor T-cell proliferation, (ii) decreased Th-1 cytokine (tumor necrosis factor-α, IFN-γ) production and (iii) increased Th-2 cytokine (IL-10, IL-4) production. In contrast to pre-treating donor mice, administering atorvastatin to recipient mice only, also protected against acute GVHD with a long-term survival of 50% compared with 0% survival in control animals. APCs of recipient mice pretreated with atorvastatin or placebo were analyzed at different time points after transplantation. Surface expression of CD80, CD86, CD40, and MHC class II antigens were decreased in animals treated with the atorvastatin as compared with the placebo group. Interestingly pre-treating both donor and recipient mice with atorvastatin produced synergistic protective effects (65% survival), when compared with survival of only donor or recipient pretreated groups (approximately 40% survival). Perhaps the most significant finding was that atorvastatin pretreatment did not interfere with perforin-mediated cytolysis or Fas-Fas-ligand interaction-mediated anti-leukemia effects.

The M.D. Anderson group has also shown prevention of acute GVHD by lovastatin mediated inhibition of LFA-1 (leukocyte function-associated antigen-1) activation, thereby preventing donor T-cells from homing to lymph nodes and Peyer's patches, in a murine BMT model [31]. Yoon et al. recently evaluated the role of statins in preventing chronic GVHD in a B10.D2→BALB/c murine BMT model [32]. The onset of clinical cutaneous chronic GVHD was significantly slower in pravastatin-treated mice compared to the control group (36 days vs. 25 days, respectively, P<0.05). Animals injected with pravastatin also had less submucosal fibrosis in lungs. Pravastatin also significantly reduced protein and chemokine concentrations and number of inflammatory cells in bronchoal-veolar lavage fluid. Significantly lower numbers of donor CD11b and CD4 cells were observed in skin and bronchoalveolar lavage fluid after pravastatin treatment. This study suggests that the chronic GVHD protecting effect of statins may involve down-regulation of chemokine production and reduction of collagen synthesis.

In humans, increased Th-2 cytokine levels before allogeneic HSCT are associated with less acute GVHD and improved TRM [33-35]. Ator-vastatin has been shown to induce Th-2 polarization when administered to healthy volunteers or patients with acute coronary syndrome or autoimmune disorders [36-39]. Interestingly co-administration of atorvastatin and cyclosporine (an agent commonly used for GVHD prophylaxis in human) in dogs have shown synergistic inhibitory effects on mitochondrial function in activated T-cells [40]. These clinical observations highlight the fact that the reduced acute GVHD seen in murine models [30] by atorvastatin induced Th-2 polarization can be translated into real clinical benefit at the bedside. Moreover, the statin-induced down regulation of co-stimulatory molecules and MHC-II expression on APCs, responsible for abrogating acute GVHD in recipient mice [30], has also been demonstrated in humans treated with statin drugs [28, 37].

Statins and clinical GVHD

Prompted by these data, we retrospectively investigated whether statin use at the time of clinical allogeneic HSCT would result in reduced acute GVHD while sparing the beneficial graft-versus-leukemia (GVL) effects (Table 1) [41]. Sixty-seven consecutive patients with acute leukemia undergoing allogeneic transplantation were evaluated. Patients taking any statin drug (at a dose of ≥ 40mg/day) for at least one month before and three months after transplantation were compared to those without a history of statin use. The two groups had similar baseline characteristics. Acute GVHD was scored according to the consensus criteria.[42] The rate of grade 2-4 acute GVHD was 10% (n=1) in the statin group compared to 40% (n=23) in the control group (p-value=0.08). On subgroup analysis of patients with acute myeloid leukemia only (n=49), a significantly reduced incidence of grade 2-4 acute GVHD was seen in statin group (0%) compared to the control group (43%) (p-value=0.02). We also analyzed if statin use, while reducing acute GVHD, mitigated the GVL effects. Kaplan-Meier estimates of 3-year progression free survival (PFS) in patients with or without statin use were 54% and 28% respectively (p-value=0.17). This non significant trend of improved PFS suggested that the GVL effects are preserved in patients using statins at the time of ASCT. Rotta et al. [43] retrospectively analyzed correlation between statin use and risk of acute GVHD in 567 patients undergoing matched sibling allogeneic HSCT (Table 1). Compared to transplants where neither the donor nor the recipient were using a statin at the time of transplantation (n = 464), statin use by the donor was associated with a decreased risk of grade 3-4 acute GVHD (hazard ratio, 0.28; 95% CI, 0.1-0.9). Statin use by both the donor and recipient (n=12) was suggestively associated with a decreased risk of grade 3 or 4 acute GVHD (hazard ratio, 0.00; 95% CI, undefined). Risks of disease relapse and TRM were not significantly affected by donor or recipient statin exposure. Interestingly statin use by the recipient only, in contrast to our previous findings [41], did not confer protection against GVHD. Similarly protective effects of statins were more prominent in patients receiving cyclosporine for GVHD prophylaxis, compared to the ones receiving tacrolimus. Recently synergistic activity of atorvastatin and cyclosporine in inhibiting T-cell activation in an animal model has been reported, partially explaining this intriguing synergy between statins and cyclosporine [40]. However the animal model did not address, whether a similar synergy existed between statins and other calcineurin inhibitors.

Table 1.

Studies evaluatingthe role of statins in preventing GVHD

Author Journal Year Study Design N Acute GVHD Chronic GVHD Comments
Hamadani [41] Blood 2008 Statin use by recipients only 67 Significant reduction No effect Limited by small sample size and retrospective design Included both MS and MUD
Rotta [43] Blood 2010 Statin use by both recipients and donors 567 Significant reduction No effect Included MS only Benefit confined to patients receiving cyclosporine
Rotta [44] BBMT 2010 Statin use by recipients only 1206 Significant reduction (in MS only) Significant reduction Large, retrospective study Included both MS and MUD Suggest increased risk of disease relapse with statin use

Abbreviations: MUD=matched unrelated donors; MS=matched sibling donors.

A recent large retrospective study evaluated the role of statin use by transplant recipients only [44], in preventing GVHD. In line with our original observation, this study reported that statin use in patients undergoing matched sibling transplantation was associated with significantly reduced rates of severe acute GVHD. Of note in contrast to prior publications [41, 43], in this study statin use was also associated significantly reduced rates of extensive chronic GVHD.

In summary, extensive preclinical and retrospective clinical data suggest that statins can prevent acute (and possibly chronic) GVHD without decreasing GVL effects. In order to prospectively validate these observations, a phase-II study evaluating the role of atorvastatin in preventing acute GVHD in patients undergoing matching sibling allogeneic HSCT is currently in advanced stage of patient accrual at our cancer center (NCT01175148; www.clinicaltrials.gov). A similar study is in currently in planning stages at Ohio State University, USA (Steven Devine, M.D.; personal communication).

Anti-myeloma activity of statins

Statins have demonstrated antineoplastic and pro-apoptotic activities in various in-vitro studies involving myeloma cell lines. Van de Donk et al. [45] reported that lovastatin can inhibit membrane-localization and resultant cytosolic accumulation of Ras by inhibition of isoprenylation, leading to markedly decreased viability of plasma cells. The cytotoxic effect of lovastatin on plasma cells is possibly the consequence of interference with the isoprenylation of Ras and its subsequent involvement in signaling cascades thereby inhibiting proliferation and cell survival signals. HMG-CoA reductase is differentially up-regulated in cell adhesion-mediated drug resistant (CAM-DR) and de novo resistant myeloma cell lines [46]. Interestingly co-cultering statins (simvastatin and lovastatin) with melphalan, resistant myeloma cells, and bone marrow stromal cells leads to complete abrogation of CAM-DR [47]. Further experiments with specific inhibitors of components of the HMG-CoA pathway revealed that CAM-DR in multiple myeloma seems to be mediated by activation of the HMG-CoA/Geranylgeranyl Pyro-phosphate/Rho-protein/Rho-kinase pathway. Microarray analyses also demonstrated that de novo and acquired drug resistance in myeloma cell lines was associated with an increase of HMG-CoA reductase gene expression [47]. In a different study, statins co-cultured with myeloma cell lines reduced mitochondrial membrane potential and inhibited the cytosolic release of mitochondria-derived activator of cas-pases, leading to activation of caspases 9, 3, 8 and resultant apoptosis [48]. In a small study, addition of simvastatin to bortezomib or benda-mustine in a total of 6 patients with refractory myeloma was shown to produce an apparent improvement in response [49].

Role of statins in autologous transplantation for myeloma

Based on the preliminary evidence that statins may serve as sensitizing agents for other che-motherapeutic agents in multiple myeloma, we evaluated the impact of statin use by myeloma patients at the time of autologous HSCT, on transplantation outcomes [50]. In a cohort of one hundred and forty-six consecutive patients with multiple myeloma undergoing autologous HSCT, outcomes of patients taking statins for at least one month before and after transplant (n=28) were compared to those without a history of statin use (n=118). The two groups had similar baseline characteristics including age, international stage, number of prior therapies, and percentage of patients with high-risk cytogenetics. A trend towards higher overall response rate post-transplant was seen in the statin group (93% vs. 78%; p=0.07). In patients with high-risk cytogenetics, the overall response rate in the statin group (n=5) was 100% versus 78% for non-statin group (n=14). This better response rate, however did not translate into improved overall survival. It is possible the sample size of the study limited the detection of a statistically significant difference between the two groups. The excellent therapeutic index of statins and their potential anti-myeloma activity, warrant further investigation of these agents in myeloma patients undergoing autologous HSCT.

Statins as modulators of bronchiolitis obliterans

Bronchiolitis obliterans is a well-recognized non-infectious, late-onset pulmonary complication of allogeneic HSCT, and is associated with substantial morbidity and mortality [51]. Bronchiolitis obliterans typically presents with symptoms of cough, dyspnea and/or airway obstruction. Pathologically, it results from obstruction and/or obliteration of the small airways and is characterized by luminal occlusion of the terminal and respiratory bronchioles by inflammatory and fibrous tissue [51]. Data from lung transplants suggest that statin use by transplant recipients is associated with significantly reduced rates of acute organ rejection, obliterative bronchiolitis and a superior overall survival [52]. These substantial clinical benefits associated with statin use after pulmonary transplantation, may have clinical significance in reducing similar pulmonary complications following allogeneic HSCT. Duncan et al. [53] reported on the safety and tolerability of statins (pravastatin) in pediatric allogeneic HSCT patients with established bronchiolitis obliterans. All participants tolerated the drug without difficulty, with evidence of clinical improvement in pulmonary function and there were no statin-associated adverse effects. Changes in creatine kinase (CK) and transaminases were minimal. One patient experienced increased CK and alanine aminotransferase, and a decrease in platelet count in the setting of severe systemic illness. Statins merit further study in this significant complication of allografting.

Conclusions

The pleiotropic immunomodulatory effects of statins and their excellent safety profile, make them an ideal yet inexpensive agent, well suited for further study in improving HSCT outcomes. The ongoing phase II clinical trials, will clarify their role in preventing acute GVHD following matched sibling allogeneic HSCT (NCT01175148; www.clinicaltrials.gov). If activity of statins in preventing transplant related complications is confirmed in well designed prospective studies, it will have significant effect on offsetting tremendous costs associated with current available strategies for preventing and treating transplant complications.

Acknowledgments

This work is supported in part by research funding by American Cancer Society 116837-IRG-09 -061-01 (M.H) and ASBMT & Millennium New Investigator Award (M.H).

References

  • 1.Hamadani M, Awan FT, Copelan EA. Hematopoietic stem cell transplantation in adults with acute myeloid leukemia. Biol Blood Marrow Transplant. 2008;14:556–567. doi: 10.1016/j.bbmt.2008.02.019. [DOI] [PubMed] [Google Scholar]
  • 2.Deeg HJ. How I treat refractory acute GVHD. Blood. 2007;109:4119–4126. doi: 10.1182/blood-2006-12-041889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Liao JK, Laufs U. Pleiotropic effects of statins. Annu Rev Pharmacol Toxicol. 2005;45:89–118. doi: 10.1146/annurev.pharmtox.45.120403.095748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart JC, Gotto AM, Greten H, Kastelein JJ, Shepherd J, Wenger NK, Treating to New Targets (TNT) Investigators Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med. 2005;352:1425–1435. doi: 10.1056/NEJMoa050461. [DOI] [PubMed] [Google Scholar]
  • 5.Wenger NK, Lewis SJ, Herrington DM, Bittner V, Welty FK, Treating to New Targets Study Steering Committee and Investigators Outcomes of using high- or low-dose atorvastatin in patients 65 years of age or older with stable coronary heart disease. Ann Intern Med. 2007;147:1–9. doi: 10.7326/0003-4819-147-1-200707030-00002. [DOI] [PubMed] [Google Scholar]
  • 6.Deedwania P, Barter P, Carmena R, Fruchart JC, Grundy SM, Haffner S, Kastelein JJ, LaRosa JC, Schachner H, Shepherd J, Waters DD, Treating to New Targets Investigators Reduction of low-density lipoprotein cholesterol in patients with coronary heart disease and metabolic syndrome: analysis of the Treating to New Targets study. Lancet. 2006;368:919–928. doi: 10.1016/S0140-6736(06)69292-1. [DOI] [PubMed] [Google Scholar]
  • 7.Newman CB, Palmer G, Silbershatz H, Szarek M. Safety of atorvastatin derived from analysis of 44 completed trials in 9,416 patients. Am J Cardiol. 2003;92:670–676. doi: 10.1016/s0002-9149(03)00820-8. [DOI] [PubMed] [Google Scholar]
  • 8.Nash RA, Antin JH, Karanes C, Fay JW, Avalos BR, Yeager AM, Przepiorka D, Davies S, Petersen FB, Bartels P, Buell D, Fitzsimmons W, Anasetti C, Storb R, Ratanatharathorn V. Phase 3 study comparing methotrexate and tacrolimus with methotrexate and cyclosporine for prophylaxis of acute graft-versus-host disease after marrow transplantation from unrelated donors. Blood. 2000;96:2062–2068. [PubMed] [Google Scholar]
  • 9.Ratanatharathorn V, Nash RA, Przepiorka D, Devine SM, Klein JL, Weisdorf D, Fay JW, Nademanee A, Antin JH, Christiansen NP, van der Jagt R, Herzig RH, Litzow MR, Wolff SN, Longo WL, Petersen FB, Karanes C, Avalos B, Storb R, Buell DN, Maher RM, Fitzsimmons WE, Wingard JR. Phase III study comparing methotrexate and tacrolimus (prograf, FK506) with methotrexate and cyclosporine for graft-versus-host disease prophylaxis after HLA-identical sibling bone marrow transplantation. Blood. 1998;92:2303–2314. [PubMed] [Google Scholar]
  • 10.Morris EC, Rebello P, Thomson KJ, Peggs KS, Kyriakou C, Goldstone AH, Mackinnon S, Hale G. Pharmacokinetics of alemtuzumab used for in vivo and in vitro T-cell depletion in allogeneic transplantations: relevance for early adoptive immunotherapy and infectious complications. Blood. 2003;102:404–406. doi: 10.1182/blood-2002-09-2687. [DOI] [PubMed] [Google Scholar]
  • 11.Hamadani M, Hofmeister CC, Jansak B, Phillips G, Elder P, Blum W, Penza S, Lin TS, Klisovic R, Marcucci G, Farag SS, Devine SM. Addition of infliximab to standard acute graft-versus-host disease prophylaxis following allogeneic peripheral blood cell transplantation. Biol Blood Marrow Transplant. 2008;14:783–789. doi: 10.1016/j.bbmt.2008.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Wagner JE, Thompson JS, Carter SL, Kernan NA, Unrelated Donor Marrow Transplantation Trial Effect of graft-versus-host disease prophylaxis on 3-year disease-free survival in recipients of unrelated donor bone marrow (T-cell Depletion Trial): a multi-centre, randomised phase II-III trial. Lancet. 2005;366:733–741. doi: 10.1016/S0140-6736(05)66996-6. [DOI] [PubMed] [Google Scholar]
  • 13.Bacigalupo A, Lamparelli T, Bruzzi P, Guidi S, Alessandrino PE, di Bartolomeo P, Oneto R, Bruno B, Barbanti M, Sacchi N, Van Lint MT, Bosi A. Antithymocyte globulin for graft-versus-host disease prophylaxis in transplants from unrelated donors: 2 randomized studies from Gruppo Italiano Trapianti Midollo Osseo (GITMO) Blood. 2001;98:2942–2947. doi: 10.1182/blood.v98.10.2942. [DOI] [PubMed] [Google Scholar]
  • 14.de Lissovoy G, Hurd D, Carter S, Beatty P, Ewell M, Henslee-Downey J, Kernan N, Yanovich S, Weisdorf D. Economic analysis of unrelated allogeneic bone marrow transplantation: results from the randomized clinical trial of T-cell depletion vs unmanipulated grafts for the prevention of graft-versus-host disease. Bone Marrow Transplant. 2005;36:539–546. doi: 10.1038/sj.bmt.1705078. [DOI] [PubMed] [Google Scholar]
  • 15.Lee SJ, Klar N, Weeks JC, Antin JH. Predicting costs of stem-cell transplantation. J Clin Oncol. 2000;18:64–71. doi: 10.1200/JCO.2000.18.1.64. [DOI] [PubMed] [Google Scholar]
  • 16.Svahn BM, Ringden O, Remberger M. Treatment costs and survival in patients with grades III-IV acute graft-versus-host disease after allogenic hematopoietic stem cell transplantation during three decades. Transplantation. 2006;81:1600–1603. doi: 10.1097/01.tp.0000210324.44633.b1. [DOI] [PubMed] [Google Scholar]
  • 17.Youssef S, Stuve O, Patarroyo JC, Ruiz PJ, Radosevich JL, Hur EM, Bravo M, Mitchell DJ, Sobel RA, Steinman L, Zamvil SS. The HMG-CoA reductase inhibitor, atorvastatin, promotes a Th2 bias and reverses paralysis in central nervous system autoimmune disease. Nature. 2002;420:78–84. doi: 10.1038/nature01158. [DOI] [PubMed] [Google Scholar]
  • 18.Leung BP, Sattar N, Crilly A, Prach M, McCarey DW, Payne H, Madhok R, Campbell C, Gracie JA, Liew FY, McInnes IB. A novel anti-inflammatory role for simvastatin in inflammatory arthritis. J Immunol. 2003;170:1524–1530. doi: 10.4049/jimmunol.170.3.1524. [DOI] [PubMed] [Google Scholar]
  • 19.Greenwood J, Steinman L, Zamvil SS. Statin therapy and autoimmune disease: from protein prenylation to immunomodulation. Nat Rev Immunol. 2006;6:358–370. doi: 10.1038/nri1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Greenwood J, Walters CE, Pryce G, Kanuga N, Beraud E, Baker D, Adamson P. Lovastatin inhibits brain endothelial cell Rho-mediated lymphocyte migration and attenuates experimental autoimmune encephalomyelitis. FASEB J. 2003;17:905–907. doi: 10.1096/fj.02-1014fje. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dunn SE, Youssef S, Goldstein MJ, Prod'homme T, Weber MS, Zamvil SS, Steinman L. Isopre-noids determine Th1/Th2 fate in pathogenic T cells, providing a mechanism of modulation of autoimmunity by atorvastatin. J Exp Med. 2006;203:401–412. doi: 10.1084/jem.20051129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fowler DH, Kurasawa K, Husebekk A, Cohen PA, Gress RE. Cells of Th2 cytokine phenotype prevent LPS-induced lethality during murine graft-versus-host reaction. Regulation of cyto-kines and CD8+ lymphoid engraftment. J Immunol. 1994;152:1004–1013. [PubMed] [Google Scholar]
  • 23.Foley JE, Mariotti J, Ryan K, Eckhaus M, Fowler DH. Th2 cell therapy of established acute graft-versus-host disease requires IL-4 and IL-10 and is abrogated by IL-2 or host-type antigen-presenting cells. Biol Blood Marrow Transplant. 2008;14:959–972. doi: 10.1016/j.bbmt.2008.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Krenger W, Snyder KM, Byon JC, Falzarano G, Ferrara JL. Polarized type 2 alloreactive CD4+ and CD8+ donor T cells fail to induce experimental acute graft-versus-host disease. J Immunol. 1995;155:585–593. [PubMed] [Google Scholar]
  • 25.Mausner-Fainberg K, Luboshits G, Mor A, Maysel-Auslender S, Rubinstein A, Keren G, George J. The effect of HMG-CoA reductase inhibitors on naturally occurring CD4+CD25+ T cells. Atherosclerosis. 2008;197:829–839. doi: 10.1016/j.atherosclerosis.2007.07.031. [DOI] [PubMed] [Google Scholar]
  • 26.Mira E, Leon B, Barber DF, Jimenez-Baranda S, Goya I, Almonacid L, Marquez G, Zaballos A, Martinez-A C, Stein JV, Ardavin C, Manes S. Statins induce regulatory T cell recruitment via a CCL1 dependent pathway. J Immunol. 2008;181:3524–3534. doi: 10.4049/jimmunol.181.5.3524. [DOI] [PubMed] [Google Scholar]
  • 27.Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, Strober S, Negrin RS. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003;9:1144–1150. doi: 10.1038/nm915. [DOI] [PubMed] [Google Scholar]
  • 28.Shimabukuro-Vornhagen A, Liebig T, von Bergwelt-Baildon M. Statins inhibit human APC function: implications for the treatment of GVHD. Blood. 2008;112:1544–1545. doi: 10.1182/blood-2008-04-149609. [DOI] [PubMed] [Google Scholar]
  • 29.Shlomchik WD, Couzens MS, Tang CB, McNiff J, Robert ME, Liu J, Shlomchik MJ, Emerson SG. Prevention of graft versus host disease by inactivation of host antigen-presenting cells. Science. 1999;285:412–415. doi: 10.1126/science.285.5426.412. [DOI] [PubMed] [Google Scholar]
  • 30.Zeiser R, Youssef S, Baker J, Kambham N, Steinman L, Negrin RS. Preemptive HMG-CoA reductase inhibition provides graft-versus-host disease protection by Th-2 polarization while sparing graft-versus-leukemia activity. Blood. 2007;110:4588–4598. doi: 10.1182/blood-2007-08-106005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang Y, Li D, Jones D, Bassett R, Sale GE, Khalili J, Komanduri KV, Couriel DR, Champlin RE, Molldrem JJ, Ma Q. Blocking LFA-1 activation with lovastatin prevents graft-versus-host disease in mouse bone marrow transplantation. Biol Blood Marrow Transplant. 2009;15:1513–1522. doi: 10.1016/j.bbmt.2009.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yoon HK, Lim JY, Kim TJ, Cho CS, Min CK. Effects of pravastatin on murine chronic graft-versus-host disease. Transplantation. 2010;90:853–860. doi: 10.1097/TP.0b013e3181f2c92b. [DOI] [PubMed] [Google Scholar]
  • 33.Lin MT, Storer B, Martin PJ, Tseng LH, Gooley T, Chen PJ, Hansen JA. Relation of an interleukin-10 promoter polymorphism to graft-versus-host disease and survival after hematopoietic-cell transplantation. N Engl J Med. 2003;349:2201–2210. doi: 10.1056/NEJMoa022060. [DOI] [PubMed] [Google Scholar]
  • 34.Holler E, Roncarolo MG, Hintermeier-Knabe R, Eissner G, Ertl B, Schulz U, Knabe H, Kolb HJ, Andreesen R, Wilmanns W. Prognostic significance of increased IL-10 production in patients prior to allogeneic bone marrow transplantation. Bone Marrow Transplant. 2000;25:237–241. doi: 10.1038/sj.bmt.1702126. [DOI] [PubMed] [Google Scholar]
  • 35.Baker KS, Roncarolo MG, Peters C, Bigler M, DeFor T, Blazar BR. High spontaneous IL-10 production in unrelated bone marrow transplant recipients is associated with fewer transplant-related complications and early deaths. Bone Marrow Transplant. 1999;23:1123–1129. doi: 10.1038/sj.bmt.1701780. [DOI] [PubMed] [Google Scholar]
  • 36.Kanda H, Yokota K, Kohno C, Sawada T, Sato K, Yamaguchi M, Komagata Y, Shimada K, Yamamoto K, Mimura T. Effects of low-dosage simvastatin on rheumatoid arthritis through reduction of Th1/Th2 and CD4/CD8 ratios. Mod Rheumatol. 2007;17:364–368. doi: 10.1007/s10165-007-0589-4. [DOI] [PubMed] [Google Scholar]
  • 37.Peng X, Jin J, Giri S, Montes M, Sujkowski D, Tang Y, Smrtka J, Vollmer T, Singh I, Markovic-Plese S. Immunomodulatory effects of 3-hydroxy-3-methylglutaryl coenzyme-A reductase inhibitors, potential therapy for relapsing remitting multiple sclerosis. J Neuroimmunol. 2006;178:130–139. doi: 10.1016/j.jneuroim.2006.06.005. [DOI] [PubMed] [Google Scholar]
  • 38.Shimada K, Miyauchi K, Daida H. Early intervention with atorvastatin modulates TH1/TH2 imbalance in patients with acute coronary syndrome: from bedside to bench. Circulation. 2004;109:e213–4. doi: 10.1161/01.CIR.0000127616.70152.5D. author reply e213-4. [DOI] [PubMed] [Google Scholar]
  • 39.Sellner J, Greeve I, Findling O, Kamm CP, Minten C, Engelhardt B, Grandgirard D, Leib SL, Mattle HP. Effect of interferon-beta and atorvastatin on Th1/Th2 cytokines in multiple sclerosis. Neurochem Int. 2008;53:17–21. doi: 10.1016/j.neuint.2008.04.004. [DOI] [PubMed] [Google Scholar]
  • 40.Margineantu DH, Hockenbery D, Martin PJ, Storb R. Statins Synergize With Cyclosporine in Inhibiting Mitochondrial Function in Activated T Cells. Biol Blood Marrow Transplant. 2011;17:S166. [Google Scholar]
  • 41.Hamadani M, Awan FT, Devine SM. The impact of HMG-CoA reductase inhibition on the incidence and severity of graft-versus-host disease in patients with acute leukemia undergoing allogeneic transplantation. Blood. 2008;111:3901–3902. doi: 10.1182/blood-2008-01-132050. [DOI] [PubMed] [Google Scholar]
  • 42.Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J, Thomas ED. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995;15:825–828. [PubMed] [Google Scholar]
  • 43.Rotta M, Storer BE, Storb RF, Martin PJ, Heimfeld S, Peffer A, Maloney DG, Deeg HJ, Sandmaier BM, Appelbaum FR, Mielcarek M. Donor statin treatment protects against severe acute graft-versus-host disease after related allogeneic hematopoietic cell transplantation. Blood. 2010;115:1288–1295. doi: 10.1182/blood-2009-08-240358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rotta M, Storer BE, Storb R, Martin PJ, Flowers ME, Vernon MS, Peffer A, Maloney DG, Deeg HJ, Sandmaier BM, Appelbaum FR, Mielcarek M. Impact of recipient statin treatment on graft-versus-host disease after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2010;16:1463–1466. doi: 10.1016/j.bbmt.2010.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.van de Donk NW, Kamphuis MM, Lokhorst HM, Bloem AC. The cholesterol lowering drug lovastatin induces cell death in myeloma plasma cells. Leukemia. 2002;16:1362–1371. doi: 10.1038/sj.leu.2402501. [DOI] [PubMed] [Google Scholar]
  • 46.Hazlehurst LA, Enkemann SA, Beam CA, Argilagos RF, Painter J, Shain KH, Saporta S, Boulware D, Moscinski L, Alsina M, Dalton WS. Genotypic and phenotypic comparisons of de novo and acquired melphalan resistance in an isogenic multiple myeloma cell line model. Cancer Res. 2003;63:7900–7906. [PubMed] [Google Scholar]
  • 47.Schmidmaier R, Baumann P, Simsek M, Dayyani F, Emmerich B, Meinhardt G. The HMG-CoA reductase inhibitor simvastatin overcomes cell adhesion-mediated drug resistance in multiple myeloma by geranylgeranylation of Rho protein and activation of Rho kinase. Blood. 2004;104:1825–1832. doi: 10.1182/blood-2003-12-4218. [DOI] [PubMed] [Google Scholar]
  • 48.Cafforio P, Dammacco F, Gernone A, Silvestris F. Statins activate the mitochondrial pathway of apoptosis in human lymphoblasts and myeloma cells. Carcinogenesis. 2005;26:883–891. doi: 10.1093/carcin/bgi036. [DOI] [PubMed] [Google Scholar]
  • 49.Schmidmaier R, Baumann P, Bumeder I, Meinhardt G, Straka C, Emmerich B. First clinical experience with simvastatin to overcome drug resistance in refractory multiple myeloma. Eur J Haematol. 2007;79:240–243. doi: 10.1111/j.1600-0609.2007.00902.x. [DOI] [PubMed] [Google Scholar]
  • 50.Hamadani M, Hade E, Benson DM, Jr, Hofmeister CC. The effect of statin use at the time of autologous transplant on response and survival in multiple myeloma. Biol Blood Marrow Transplant. 2008;14:351–352. doi: 10.1016/j.bbmt.2007.12.489. [DOI] [PubMed] [Google Scholar]
  • 51.Duncan CN, Buonanno MR, Barry EV, Myers K, Peritz D, Lehmann L. Bronchiolitis obliterans following pediatric allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2008;41:971–975. doi: 10.1038/bmt.2008.19. [DOI] [PubMed] [Google Scholar]
  • 52.Johnson BA, Iacono AT, Zeevi A, McCurry KR, Duncan SR. Statin use is associated with improved function and survival of lung allografts. Am J Respir Crit Care Med. 2003;167:1271–1278. doi: 10.1164/rccm.200205-410OC. [DOI] [PubMed] [Google Scholar]
  • 53.Duncan CN, Barry EV, Lehmann LE. Tolerability of pravastatin in pediatric hematopoietic stem cell transplant patients with bronchiolitis obliterans. J Pediatr Hematol Oncol. 2010;32:185–188. doi: 10.1097/MPH.0b013e3181d32184. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Blood Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES