Abstract
Hydroxymethylglutaryl Co-enzyme A reductase inhibitors, also known as statins, are a class of anti-hyperlipidemic agents. These drugs have been employed vastly to reduce the morbidity and mortality of cardiovascular disorders. Soon after their introduction, benefits other than their primary actions were discovered. Along with these pleiotropic properties, a series of mainly favorable effects has been proposed in patients intended to undergo hematopoietic stem cell transplantation. These actions address some complications encountered by this special population such as graft-versus-host disease, efficacy of chemotherapy, infections, etc. This review presents the current evidence surrounding these issues.
Key Words: Infection, GvHD, Statin, Stem cell transplantation
Introduction
Inhibitors of 3-hydroxy3-methylglutaryl Coenzyme A reductase or statins were developed primarily to reduce the risk of cardiovascular diseases. They were used later to address several new indications unrelated to the primary lipid-lowering effect. Among them, new promising roles have been proposed in hematopoietic stem cell transplantation (HSCT). 1
Although HSCT is a life-saving approach for various types of hematological malignancies, it imposes noticeable treatment-related morbidity and mortality on patients. 2-4 A number of recently published studies have evaluated the possible advantageous roles of statins in some complications encountered after HSCT. Our focus has been more on Graft vs. Host Disease (GvHD), infection risk, efficacy of chemotherapy, and hypercholesterolemia. Here, we will scrutinize these emerging proceedings.
Statins and the risk of GvHD
GvHD is one of the main challenges of HSCT survivors. Both animal and human studies implicating various statins have demonstrated the effectiveness of these agents in this setting; 5,6 but depending on whether the drug had been administered to the donor or the recipient, inconsistent results were obtained. Rotta et al. evaluated transplant outcomes of 1206 patients undergoing allogeneic HSCT retrospectively. 5 Donors were either HLA-identical siblings or HLA-matched unrelated ones. Recipients with a history of statin use for at least 3 months prior to transplantation were considered as “on statin treatment”. Results were adjusted for age, gender, donor type, conditioning intensity, source of stem cells, and specific calcineurin inhibitor (CNI) agent used for GvHD prophylaxis. There were no differences in grade II-IV acute GvHD (aGvHD), non-relapse mortality and overall mortality between statin users and non-users. However, less extensive chronic GvHD (cGvHD) occurred in recipients previously treated with statin. The latter effect was observed in patients treated with cyclosporine only and not in those who received tacrolimus as GvHD prophylaxis. This effect also was associated with more frequent recurrence of underlying malignancy. The same authors evaluated the effect of donor statin use on the incidence of GvHD in another retrospective study. 7
Five hundred and sixty-seven patients were included. After adjustment for other variables in multivariable analysis, donor statin use was associated with reduced rate of grade III-IV but not grade II-IV aGvHD. Again, this effect was observed only in patients who were prescribed cyclosporine as post-engraftment prophylaxis. There was no difference between the two groups in terms of cGvHD. After further examination of involved organs, more protective effects were observed in the gastrointestinal tract involvement and not in the skin. Similar results have been obtained in a trial conducted by Hamadani et al.
In this phase II study, simultaneous donor and recipient pretreatment with atorvastatin 40 mg daily orally for 14-28 days before cell harvest was well tolerated and resulted in reduced incidence of acute GvHD. 8 Of note, donors in this study were matched siblings and recipients were treated with tacrolimus as the immunosuppressant agent of choice. In general and based on limited evidence available, it can be hypothesized that donor statin treatment is associated with reduced incidence of aGvHD, while recipient pretreatment with statin may cause less severe cGvHD. More prospective randomized trials are needed to further scrutinize these preliminary results.
Besides HSCT, immunosuppressive therapy is needed in solid organ transplantation and some autoimmune disorders. Statins also seem promising as new add-on therapies in these states which will be discussed later in this article.
Statins and the risk of infection in post-transplantation period
There is not sufficient evidence available about the effects of these drugs on the risk of infections after HSCT. One study used the same database as one of the articles mentioned above. 9 In this retrospective analysis, neither the donor nor the recipient’s statin use was found to reduce the risk of some infections. These included CMV infection, bacteremia, invasive fungal infections and lower respiratory tract infections. Those recipients who were taking statins; however, experienced more cumulative episodes of gram-negative bacteremia that did not result in increased mortality. Donor’s statin use was also associated with increased viral respiratory tract infections.
Due to the retrospective design of this study and also due to the lack of prospective trials, it is very difficult to conclude any beneficial or detrimental effect of statin use on the risk of infections in this population.
Statin use to enhance chemotherapy efficacy in potential transplantation candidates
Proliferation of multiple myeloma (MM) cells is inhibited in the presence of statins. It has been hypothesized that statin exposure leads in release of some caspases (namely caspase 9, 3, and 8) with subsequent apoptosis of MM cells. 10 Not all MM cell lines are sensitive to statins. 11 In preliminary clinical studies, these drugs appeared to be promising anti-myeloma agents. In a phase II pilot study, simultaneous simvastatin could prevent drug resistance in MM cases resistant to two cycles of bortezomib or bendamustine. 12 Enrolled patients were six post-autologous HSCT cases who received two cycles of bortezomib or bendamustine with no response.
In this case, the patients received two additional cycles plus simvastatin 80 mg daily. Simvastatin was administered from day -2 to two days after completing chemotherapy. Results were compared with another group that included 10 patients with resistant MM after 4 cycles of bortezomib or bendamustine, but without concomitant simvastatin. In 3 out of 6 patients, serum level of M protein was increasing even after two cycles of chemotherapy, but when simvastatin was added to the ongoing regimen, protein levels markedly declined. In two other patients, stable serum M protein levels began to fall consistent with the initiation of simvastatin. In only 1 patient, protein level was not affected by simvastatin treatment. In this patient; however, serum level of cholesterol increased slightly in a paradoxical manner that indicated insufficient HMG-CoA reductase inhibition. The only important limitation of this study was the small number of samples.
In another phase II trial, contradictory results were obtained with simvastatin in relapsed or refractory multiple myeloma patients. 13 Simvastatin 15 mg/kg/day divided as twice daily was added to chemotherapy regimen in days 1-7 of each cycle. Twenty-eight day cycles consisted of vincristine 0.4 mg, doxorubicin 9 mg/m2 and dexamethasone 40 mg orally on day 7-10. Response evaluation was performed after two cycles of treatment and in the case of stability; treatment was continued for two more cycles.
Disease progression after two cycles was the factor that led to discontinuation of treatment. Interim analysis was performed after recruitment of 12 patients. Initial assessment showed that one patient achieved a partial response, 6 patients were stable and other 5 patients progressed. Five out of 6 patients with stable disease were those with progressive disease before enrollment. Stability was maintained for 94-258 days (with a median of 103 days). According to the authors’ opinion, this rate of response was not noticeable enough to justify completion of the study and it was discontinued prematurely.
In a different survey, the effect of statin use at the time of autologous HSCT on transplantation outcomes was explored. 10 Hamadani et al. retrospectively analyzed the records of 146 patients undergoing high dose chemotherapy with melphalan (200 mg/m2) followed by autologous HSCT for multiple myeloma. Statin use was defined as taking any statin agent with doses ≥ 20 mg per day from 1 month before to 1 month after transplantation. Twenty-eight statin-users were compared with 118 patients who did not take statin. Complete response (CR) and very good partial response (VGPR) did not differ significantly between groups (43% vs. 45%, p=0.84), but the composite endpoint of overall response rate (CR + VGPR + PR) trended more favorable with statin use versus non-use (97% vs. 78%, p=0.07). Median overall survival (25.7 vs. 22 months, p=0.65) and progression-free survival (19.5 vs. 14.8 months, p=0.97) were not significantly different between groups. Based on the available body of evidence, it is not clear whether co-administration of statin with chemotherapy will result in enhanced efficacy or not. Well-designed randomized trials with large sample sizes are needed to better investigate this capacity.
Statin use to treat hypercholesterolemia after transplantation
With improving survival of post-HSCT patients; however, concerns have been raised about long-term non-relapse mortality. Cardiovascular disorders comprise a major constituent of a large variety of long-term complications. 14-17 In a retrospective analysis carried out by Kagoya et al. risk factors, prevalence, and prognosis of hypercholesterolemia in patients after allogeneic HSCT were examined. 18
Medical records of 194 patients who survived more than 100 days after transplantation came into the analysis. Hypercholesterolemia (defined as cholesterol levels above 240 mg/dL in at least 2 consecutive measurements one week apart) occurred in 83 of 194 patients (42.8%). Hypertriglyceridemia (defined as triglyceride levels beyond 200 mg/dL in at least two occasions one week apart) developed in 99 patients (50.8%). These abnormalities led to the administration of statins to 19, fibrate to 2, combined statin-fibrate to one, and nicotinic acid to one patient. Non-relapse mortality did not differ significantly between patients with or without hypercholesterolemia (6-year mortality 17.8% vs. 18.7% respectively; p=0.83).
In contrast to some other studies,19,20 use of CNIs was not an independent risk factor for either hypercholesterolemia or hypertriglyceridemia. Nonetheless, both cGVHD and subsequent corticosteroid use were associated with hypercholesterolemia. Statins have been previously used to treat lipid abnormalities in solid organ transplantations. 21,22 Lipid abnormalities have been successfully treated with statins. In those who did not receive cholesterol-lowering therapy, normalization of values occurred shortly after HSCT, signifying the impact of timely approach to cGvHD on lipid profile values. Another unexpected finding in the study was the lower rate of relapse of primary disease in patients with hypercholesterolemia. Nonetheless, this finding can be attributed to liver involvement due to cGvHD and ensuing graft versus leukemia effect.
In one of the largest studies conducted to date, Blaster et al. followed 1493 patients who underwent allogeneic HSCT and survived more than 100 days after transplantation. 23 Of whom, 732 patients never had done tests to measure cholesterol level and were subsequently excluded from the study. Both hypercholesterolemia and hypertriglyceridemia were defined as any single outpatient value of ≥ 200 mg/dL.
All patients with a prescription for statin for at least 30 days after the procedure were considered as statin user. Ninety-five percent of patients received tacrolimus as the CNI of choice for GvHD prophylaxis. Sirolimus was prescribed for 50% of patients. The cumulative incidence of aGvHD and cGvHD was 26% and 60% at 2 years post-transplant, respectively. Out of the 761 patients, 556 had at least one cholesterol level equals to or greater than 200 mg/dL and were considered hypercholesterolemic according to the National Cholesterol Education Panel ATP-III guideline. Out of the 761 patients, 560 had at least one lipid value measurement before transplantation which revealed 32.0% (n=179) prevalence of dyslipidemia prior to transplantation. A high proportion of patients also suffered from post-transplantation hypertriglyceridemia (n=531, 72.5%). Among patients with both pre- and post-transplant triglyceride values, the mean peak values were 171 and 275 mg/dL, respectively (a mean alteration of 109 mg/dL, p< 0.0001). In terms of the impact of transplantation procedure on lipid values, 249 of 381 patients (65%) with both pre- and post-procedural measurements, developed newly-diagnosed abnormal lipid values after transplantation. Given this rate of de novo conversion, it is noteworthy to say that 164 of 179 previously hypercholesterolemic patients remained unchanged after procedure. Patients with pre-existing hypercholesterolemia had significantly higher cholesterol levels compared with de novo cases (259 mg/dL vs. 275 mg/dL, p=0.004 respectively). Similar results were also observed in triglyceride status. Approximately, 64% of patients with pre-transplant TG level <200 mg/dL developed elevated TG levels after the procedure. Again, pre-existing hypertriglyceridemic patients exerted higher TG levels after transplantation compared with de novo subjects (398 vs. 305 mg/dL, p< 0.001). Suffering from higher grades of aGvHD was associated with higher risk for abnormal cholesterol levels (frequency of 81% for aGvHD grade II-IV vs. 71% for aGvHD grade 0-I, p=0.007). These abnormal results of lipid values led to the prescription of statins to 220 of 761 patients (29%) within 2 years after transplantation. Adverse drug reactions attributed to statin were negligible in only one case after drug discontinuation. Results of other trials unrelated to the HSCT population have also reported new favorable effects. In all of these situations, statins have been used as an adjunct to immunosuppressive regimens and have improved some indices of morbidity or mortality.
Statins have prolonged the function of allografts and improved survival in cardiac transplant patients. 24-27 Beneficial effects in lung transplantation 28 and some autoimmune disorders such as rheumatoid arthritis and multiple sclerosis have also been reported. 29-33 Together, these data suggest that statin use after HSCT not only can normalize lipid values but also may enhance immune tolerance.
Future look at statins in HSCT
Regarding the positive effects of statins after myocardial infarction, hopes for new indications of these drugs have increased. At least a part of these positive effects has been attributed to the mobilization of endothelial progenitor cells from bone marrow. 34-36 Whether similar effects on hematopoietic progenitor cells may really exist remains to be determined in future trials, but a retrospective trial showed some positive results. 37 In this study, 86 patients with MM were retrospectively analyzed. All patients received G-CSF for stem cell mobilization. The outcome of leukapheresis of 20 patients on statin treatment was compared with the other 66 non-users. The chance of adequate cell harvest with first leukapheresis was marginally superior in statin users (85% vs. 63.6%, p=0.07). This may be the subject of future trials since poor mobilizations is a barrier to successful transplantation in autologous HSCT. 38 A summary of evidence reviewed in this article is shown in Table 1.
Table 1.
Study | Design | Year | Number of patient | Comparison | Main results | Comments |
GvHD | ||||||
Rotta et al. | Retrospective | 2010 | 1206 | Recipients with statin use vs. non-users | Less extensive cGvHD in statin users | No difference in grade II-IV aGvHD, Positive results only in patients taking cyclosporine |
Rotta et al. | Retrospective | 2010 | 567 | Donor with statin use vs. non-use | Less grade III-IV aGvHD | No difference in cGvHD, Positive effects only in patients taking cyclosporine |
Hamadani et al. | Prospective | 2013 | 30 | Atorvastatin use by both donor and recipient vs. none | Less aGvHD | Positive effects in patient on tacrolimus treatment |
Infection | ||||||
Seo et al. | Retrospective | 2013 | 1206 | Recipient or donor use of statin vs. non-use | No appreciable effect on the risk of infections | Increased risk of gram-negative bacteremia with recipient statin use, Increased risk of respiratory tract viral infections with donor statin use |
Chemotherapy efficacy | ||||||
Schmidmaier et al. | Prospective Pilot Phase II | 2007 | 6 | Simvastatin added to bendamustine or bortezomib vs. bendamustine or bortezomib alone | Serum M protein declined in almost all patients after addition of simvastatin | No significant adverse effect with addition of simvastatin |
Van der Spek et al. | Prospective Phase II | 2007 | 12 | Simvastatin added to VAD vs. literature- based data for efficacy of VAD alone | No significant result at interim analysis | |
Hamadani et al. | Retrospective | 2008 | 146 | Statin use at the time of autologous HSCT for MM vs. non-use | OR trended better with statin use (not significant) | CR and VGPR did not differ significantly, A trend toward increased stem cell mobilization with statin use |
Hyperlipidemia | ||||||
Kagoya et al. | Retrospective | 2012 | 194 | - | Hypercholesterolemia in 42.8% and hypertriglyceridemia in 50.8% of patients | Successful treatment with statins |
Blaster et al. | Retrospective | 2012 | 1493 | - | Dyslipidemia in 32.0% of patients prior to transplantation, Post-procedure elevated TG in 72.5% of patients | Only one statin discontinuation because of adverse effects with 220 prescriptions |
Stem cell mobilization | ||||||
Stravodimou et al. | Retrospective | 2014 | 86 | Statin use vs. non-use in autologous HSCT for MM | A non-significant increase in stem cell mobilization |
Barriers to use of statins in post-HSCT population
Despite the aforementioned benefits, some concerns may prevent widespread use of statins. Among them, potentials for adverse drug reactions and drug interactions appear to be the most bothersome. The two major adverse effects attributed to statins are liver injury and myopathy. Liver injury is best described as an idiosyncratic reaction that is neither predictable nor dose-dependent. True incidence remains to be clarified since surrogate markers are lacking 39 and diagnosis has been traditionally based on different scoring tools that stratify the reaction based on some clinical and laboratory data. 40,41 In the case of hepatotoxicity, different approaches do exist in the literature42,43 that underscores the need for more practical tools to establish the causality. Regardless of whether this toxicity really exists or not, the true prevalence remains low. This fear should not obscure the benefits of the drug even in patients with pre-existing liver disease.44,45
Muscle toxicity is another precaution for statin use. Various definitions have been proposed by accredited authorities. 46-48 Unlike liver toxicity, this effect occurs in a dose-dependent manner. Accumulating evidence suggest that reduction in metabolites produced by mevalonate pathway is the main mechanism involved in muscle toxicity. 49
Another concern in clinical practice is the potential of these drugs to interact with some medications used by HSCT patients. Most statins are substrates of P-glycoprotein (P-gp). This protein acts as an efflux pump which drops out the drugs into the gut lumen and reduces their absorption. Through the action of transporters such as organic anion-transporting polypeptide 1B1 (OATP1B1), more statin is picked up by the liver and available for metabolism by CYP enzymes. Drugs like cyclosporine interact with several steps of statin metabolism, and cause an elevation in serum level with subsequent risk of myopathy. 50 Such interactions have also resulted in clinical cases of rhabdomyolysis. 51,52 Each agent of this class exhibits a unique metabolic pathway with different potential for drug-drug interactions. Detailed review of these pharmacokinetic properties is beyond the scope of this article and is discussed elsewhere. 50,53 In general, the well-defined risk profile of these agents has failed to prevent their widespread use for their primary indications. It seems that it is also true in the case of HSCT population.
CONCLUSION
Besides traditional lipid-lowering potential, statins have shown promising benefits in HSCT. Their primary action of controlling cholesterol level is extremely efficient because of the high prevalence of lipid disorders reported by abovementioned trials. Depending on donor or recipient statin use, varying outcomes have been observed regarding GvHD. Infections do not seem to be favorably affected by the action of these drugs. Although not thoroughly investigated, a positive effect of statins is anticipated on the efficacy of conditioning regimens prior to autologous HSCT or pre-transplantation chemotherapy of MM. The stem cell mobilizing potential needs more investigation. Nevertheless, whether these effects are agent-specific for certain statins have not been investigated. Other than their classic effect, unfortunately these new roles have been surveyed either in retrospective studies or in small-size prospective trials. More well-designed prospective trials are needed to establish new indications for these drugs.
ACKNOWLEDGEMENT
We would like to thank Dr. Motaharinia for his expert advice and encouragement through this review.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
References
- 1.Shimabukuro-Vornhagen A, Glossmann J, Liebig T, et al. The use of statins in hematopoietic stem cell transplantation. Curr Stem Cell Res Ther. 2009;4(4):260–5. [PubMed] [Google Scholar]
- 2.Dhir S, Slatter M, Skinner R. Recent advances in the management of graft-versus-host disease. Arch Dis Child. 2014;99(12):1150–7. doi: 10.1136/archdischild-2013-304832. e-pub ahead of print 12 Jul 2014; doi: 10.1136/archdischild-2013-304832. [DOI] [PubMed] [Google Scholar]
- 3.Lucena CM, Torres A, Rovira M, et al. Pulmonary complications in hematopoietic SCT: a prospective study. Bone Marrow Transplant. 2014;49(10):1293–9. doi: 10.1038/bmt.2014.151. e-pub ahead of print 21 Jul 2014; doi: 10.1038/bmt.2014.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Servais S, Lengline E, Porcher R, et al. Long-term immune reconstitution and infection burden after mismatched hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2014;20(4):507–17. doi: 10.1016/j.bbmt.2014.01.001. [DOI] [PubMed] [Google Scholar]
- 5.Rotta M, Storer BE, Storb R, et al. Impact of recipient statin treatment on graft-versus-host disease after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2010;16(10):1463–6. doi: 10.1016/j.bbmt.2010.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Yoon HK, Lim JY, Kim TJ, et al. Effects of pravastatin on murine chronic graft-versus-host disease. Transplantation. 2010 Oct 27;90(8):853–60. doi: 10.1097/TP.0b013e3181f2c92b. [DOI] [PubMed] [Google Scholar]
- 7.Rotta M, Storer BE, Storb RF, et al. Donor statin treatment protects against severe acute graft-versus-host disease after related allogeneic hematopoietic cell transplantation. Blood. 2010 Feb 11;115(6):1288–95. doi: 10.1182/blood-2009-08-240358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hamadani M, Gibson LF, Remick SC, et al. Sibling donor and recipient immune modulation with atorvastatin for the prophylaxis of acute graft-versus-host disease. J Clin Oncol. 2013 Dec 10;31(35):4416–23. doi: 10.1200/JCO.2013.50.8747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Boeckh M, Storer BE, Schubert MM, et al. The Effect Of Donor and Recipient Statin Treatment On Infections After Allogeneic Hematopoietic Cell Transplantation. Blood. 2013;122(21):4548. [Google Scholar]
- 10.Hamadani M, Hade E, Benson DM, et al. The effect of statin use at the time of autologous transplant on response and survival in multiple myeloma. Biol Blood Marrow Transplant. 2008 Mar;14(3):351–2. doi: 10.1016/j.bbmt.2007.12.489. [DOI] [PubMed] [Google Scholar]
- 11.Wong WW, Clendening JW, Martirosyan A, et al. Determinants of sensitivity to lovastatin-induced apoptosis in multiple myeloma. Mol Cancer Ther. 2007 Jun;6(6):1886–97. doi: 10.1158/1535-7163.MCT-06-0745. [DOI] [PubMed] [Google Scholar]
- 12.Schmidmaier R, Baumann P, Bumeder I, et al. First clinical experience with simvastatin to overcome drug resistance in refractory multiple myeloma. Eur J Haematol. 2007 Sep;79(3):240–3. doi: 10.1111/j.1600-0609.2007.00902.x. [DOI] [PubMed] [Google Scholar]
- 13.van der Spek E, Bloem AC, Sinnige HA, et al. High dose simvastatin does not reverse resistance to vincristine, adriamycin, and dexamethasone (VAD) in myeloma. Haematologica. 2007 Dec;92(12):e130–1. doi: 10.3324/haematol.12071. [DOI] [PubMed] [Google Scholar]
- 14.Griffith ML, Savani BN, Boord JB. Dyslipidemia after allogeneic hematopoietic stem cell transplantation: evaluation and management. Blood. 2010 Aug 26;116(8):1197–204. doi: 10.1182/blood-2010-03-276576. [DOI] [PubMed] [Google Scholar]
- 15.Tichelli A, Bucher C, Rovo A, et al. Premature cardiovascular disease after allogeneic hematopoietic stem-cell transplantation. Blood. 2007 Nov 1;110(9):3463–71. doi: 10.1182/blood-2006-10-054080. [DOI] [PubMed] [Google Scholar]
- 16.Baker KS, Gurney JG, Ness KK, et al. Late effects in survivors of chronic myeloid leukemia treated with hematopoietic cell transplantation: results from the Bone Marrow Transplant Survivor Study. Blood. 2004 Sep 15;104(6):1898–906. doi: 10.1182/blood-2004-03-1010. [DOI] [PubMed] [Google Scholar]
- 17.Savani BN, Griffith ML, Jagasia S, et al. How I treat late effects in adults after allogeneic stem cell transplantation. Blood. 2011 Mar 17;117(11):3002–9. doi: 10.1182/blood-2010-10-263095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kagoya Y, Seo S, Nannya Y, et al. Hyperlipidemia after allogeneic stem cell transplantation: prevalence, risk factors, and impact on prognosis. Clin Transplant. 2012 Mar-Apr;26(2):E168–75. doi: 10.1111/j.1399-0012.2012.01628.x. [DOI] [PubMed] [Google Scholar]
- 19.Kuster GM, Drexel H, Bleisch JA, et al. Relation of cyclosporine blood levels to adverse effects on lipoproteins. Transplantation. 1994 May 27;57(10):1479–83. [PubMed] [Google Scholar]
- 20.Satterthwaite R, Aswad S, Sunga V, et al. Incidence of new-onset hypercholesterolemia in renal transplant patients treated with FK506 or cyclosporine. Transplantation. 1998 Feb 15;65(3):446–9. doi: 10.1097/00007890-199802150-00030. [DOI] [PubMed] [Google Scholar]
- 21.Soveri I, Abedini S, Holdaas H, et al. Metabolic syndrome and cardiovascular risk in renal transplant recipients: effects of statin treatment. Clin Transplant. 2009 Nov-Dec;23(6):914–20. doi: 10.1111/j.1399-0012.2009.01025.x. [DOI] [PubMed] [Google Scholar]
- 22.Taylor PJ, Kubler PA, Lynch SV, et al. Effect of atorvastatin on cyclosporine pharmacokinetics in liver transplant recipients. Ann Pharmacother. 2004 Feb;38(2):205–8. doi: 10.1345/aph.1D388. [DOI] [PubMed] [Google Scholar]
- 23.Blaser BW, Kim HT, Alyea EP, et al. Hyperlipidemia and statin use after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2012 Apr;18(4):575–83. doi: 10.1016/j.bbmt.2011.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kobashigawa JA, Moriguchi JD, Laks H, et al. Ten-year follow-up of a randomized trial of pravastatin in heart transplant patients. J Heart Lung Transplant. 2005 Nov;24(11):1736–40. doi: 10.1016/j.healun.2005.02.009. [DOI] [PubMed] [Google Scholar]
- 25.Wenke K, Meiser B, Thiery J, et al. Simvastatin reduces graft vessel disease and mortality after heart transplantation: a four-year randomized trial. Circulation. 1997 Sep 2;96(5):1398–402. doi: 10.1161/01.cir.96.5.1398. [DOI] [PubMed] [Google Scholar]
- 26.Wenke K, Meiser B, Thiery J, et al. Simvastatin initiated early after heart transplantation: 8-year prospective experience. Circulation. 2003 Jan 7;107(1):93–7. doi: 10.1161/01.cir.0000043241.32523.ee. [DOI] [PubMed] [Google Scholar]
- 27.Mehra MR, Raval NY. Metaanalysis of statins and survival in de novo cardiac transplantation. Transplant proc. 2004 Jun;36(5):1539–41. doi: 10.1016/j.transproceed.2004.05.036. [DOI] [PubMed] [Google Scholar]
- 28.Johnson BA, Iacono AT, Zeevi A, et al. Statin use is associated with improved function and survival of lung allografts. Am J Respir Crit Care Med. 2003 May 1;167(9):1271–8. doi: 10.1164/rccm.200205-410OC. [DOI] [PubMed] [Google Scholar]
- 29.Shirinsky IV, Zheltova OI, Solovyova NY, et al. Changes in disease activity, cytokine production, and proliferation of peripheral blood mononuclear cells in patients with rheumatoid arthritis after simvastatin treatment. Scand J Rheumatol. 2009 Jan-Feb;38(1):23–7. doi: 10.1080/03009740802363776. [DOI] [PubMed] [Google Scholar]
- 30.Okamoto H, Koizumi K, Kamitsuji S, et al. Beneficial action of statins in patients with rheumatoid arthritis in a large observational cohort. J Rheumatol. 2007 May;34(5):964–8. [PubMed] [Google Scholar]
- 31.Togha M, Karvigh SA, Nabavi M, et al. Simvastatin treatment in patients with relapsing-remitting multiple sclerosis receiving interferon beta 1a: a double-blind randomized controlled trial. Mult Scler. 2010 Jul;16(7):848–54. doi: 10.1177/1352458510369147. [DOI] [PubMed] [Google Scholar]
- 32.Vollmer T, Key L, Durkalski V, et al. Oral simvastatin treatment in relapsing-remitting multiple sclerosis. Lancet. 2004 May 15;363(9421):1607–8. doi: 10.1016/S0140-6736(04)16205-3. [DOI] [PubMed] [Google Scholar]
- 33.Lanzillo R, Orefice G, Quarantelli M, et al. Atorvastatin combined to interferon to verify the efficacy (ACTIVE) in relapsing-remitting active multiple sclerosis patients: a longitudinal controlled trial of combination therapy. Mult Scler. 2010 Apr;16(4):450–4. doi: 10.1177/1352458509358909. [DOI] [PubMed] [Google Scholar]
- 34.Xu H, Yang YJ, Yang T, et al. Statins and stem cell modulation. Ageing Res Rev. 2013 Jan;12(1):1–7. doi: 10.1016/j.arr.2012.03.006. [DOI] [PubMed] [Google Scholar]
- 35.Tousoulis D, Kaski JC, Antoniades C, et al. Beneficial effects of statin treatment after myocardial infarction: Is progenitor cell mobilization the missing link? Int J Cardiol. 2008 Nov 28;130(3):301–3. doi: 10.1016/j.ijcard.2008.06.079. [DOI] [PubMed] [Google Scholar]
- 36.Leone AM, Rutella S, Giannico MB, et al. Effect of intensive vs standard statin therapy on endothelial progenitor cells and left ventricular function in patients with acute myocardial infarction: Statins for regeneration after acute myocardial infarction and PCI (STRAP) trial. Int J Cardiol. 2008 Nov 28;130(3):457–62. doi: 10.1016/j.ijcard.2008.05.036. [DOI] [PubMed] [Google Scholar]
- 37.Stravodimou A, Voutsadakis IA. Statin use and peripheral blood progenitor cells mobilization in patients with multiple myeloma. Clin Transl Oncol. 2014 Jan;16(1):85–90. doi: 10.1007/s12094-013-1046-9. [DOI] [PubMed] [Google Scholar]
- 38.Rossi G, Skert C, Morello E, et al. PBSC mobilization in lymphoma patients: analysis of risk factors for collection failure and development of a predictive score based on the kinetics of circulating CD34+ cells and WBC after chemotherapy and G-CSF mobilization. Hematol Oncol. 2015;33(3):125–32. doi: 10.1002/hon.2148. e-pub ahead of print 29 May 2014; doi: 10.1002/hon.2148. [DOI] [PubMed] [Google Scholar]
- 39.Teschke R, Frenzel C, Wolff A, et al. Rare statin hepatotoxicity: convincing evidence based on breakthrough case study. J Hepatol. 2012 Sep;57(3):699–700. doi: 10.1016/j.jhep.2012.02.031. [DOI] [PubMed] [Google Scholar]
- 40.Lee WM, Senior JR. Recognizing drug-induced liver injury: current problems, possible solutions. Toxicol Pathol. 2005;33(1):155–64. doi: 10.1080/01926230590522356. [DOI] [PubMed] [Google Scholar]
- 41.Lucena MI, Garcia-Cortes M, Cueto R, et al. Assessment of drug-induced liver injury in clinical practice. Fundam Clin Pharmacol. 2008 Apr;22(2):141–58. doi: 10.1111/j.1472-8206.2008.00566.x. [DOI] [PubMed] [Google Scholar]
- 42.Bjornsson E, Jacobsen EI, Kalaitzakis E. Hepatotoxicity associated with statins: reports of idiosyncratic liver injury post-marketing. J Hepatol. 2012 Feb;56(2):374–80. doi: 10.1016/j.jhep.2011.07.023. [DOI] [PubMed] [Google Scholar]
- 43.Bader T. The myth of statin-induced hepatotoxicity. Am J Gastroenterol. 2010 May;105(5):978–80. doi: 10.1038/ajg.2010.102. [DOI] [PubMed] [Google Scholar]
- 44.Khorashadi S, Hasson NK, Cheung RC. Incidence of statin hepatotoxicity in patients with hepatitis C. Clin Gastroenterol Hepatol. 2006 Jul;4(7):902–7. doi: 10.1016/j.cgh.2006.03.014. quiz 806. [DOI] [PubMed] [Google Scholar]
- 45.Chalasani N, Aljadhey H, Kesterson J, et al. Patients with elevated liver enzymes are not at higher risk for statin hepatotoxicity. Gastroenterology. 2004 May;126(5):1287–92. doi: 10.1053/j.gastro.2004.02.015. [DOI] [PubMed] [Google Scholar]
- 46.Pasternak RC, Smith SC, Bairey-Merz CN, et al. ACC/AHA/NHLBI Clinical Advisory on the Use and Safety of Statins. Circulation. 2002 Aug 20;106(8):1024–8. doi: 10.1161/01.cir.0000032466.44170.44. [DOI] [PubMed] [Google Scholar]
- 47.McKenney JM, Davidson MH, Jacobson TA, et al. Final conclusions and recommendations of the National Lipid Association Statin Safety Assessment Task Force. Am J Cardiol. 2006 Apr 17;97(8A):89C–94C. doi: 10.1016/j.amjcard.2006.02.030. [DOI] [PubMed] [Google Scholar]
- 48.Sewright KA, Clarkson PM, Thompson PD. Statin myopathy: incidence, risk factors, and pathophysiology. Curr Atheroscler Rep. 2007 Nov;9(5):389–96. doi: 10.1007/s11883-007-0050-3. [DOI] [PubMed] [Google Scholar]
- 49.Norata GD, Tibolla G, Catapano AL. Statins and skeletal muscles toxicity: From clinical trials to everyday practice. Pharmacol Res. 2014;88:107–13. doi: 10.1016/j.phrs.2014.04.012. e-pub ahead of print 13 May 2014; doi: 10.1016/j.phrs.2014.04.012. [DOI] [PubMed] [Google Scholar]
- 50.Kellick KA, Bottorff M, Toth PP. A clinician's guide to statin drug-drug interactions. J Clin Lipidol. 2014 May-Jun;8(3 Suppl):S30–46. doi: 10.1016/j.jacl.2014.02.010. [DOI] [PubMed] [Google Scholar]
- 51.Tong J, Laport G, Lowsky R. Rhabdomyolysis after concomitant use of cyclosporine and simvastatin in a patient transplanted for multiple myeloma. Bone marrow transplant. 2005 Oct;36(8):739–40. doi: 10.1038/sj.bmt.1705128. [DOI] [PubMed] [Google Scholar]
- 52.Shahan JL, Panu LD, Hildebrandt GC. Rhabdomyolysis in a multiple myeloma patient secondary to concurrent treatment with lenalidomide and pravastatin and to lenalidomide alone. Int J Hematol. 2012 Dec;96(6):818–9. doi: 10.1007/s12185-012-1226-3. [DOI] [PubMed] [Google Scholar]
- 53.Corsini A, Bellosta S, Baetta R, et al. New insights into the pharmacodynamic and pharmacokinetic properties of statins. Pharmacol Ther. 1999 Dec;84(3):413–28. doi: 10.1016/s0163-7258(99)00045-5. [DOI] [PubMed] [Google Scholar]