throbber
r 2009 John Wiley & Sons A/S
`
`Transplant Infectious Disease . ISSN 1398 -2273
`
`Review article
`
`Mycophenolate mofetil: e¡ects on cellular immune
`subsets, infectious complications, and antimicrobial
`activity
`
`M.L. Ritter, L. Pirofski. Mycophenolate mofetil: e¡ects on cellular
`immune subsets, infectious complications, and antimicrobial activity.
`Transpl Infect Dis 2009: 11: 290^297. All rights reserved
`
`Abstract : Mycophenolate mofetil (MMF) is one of the most frequently
`used immunosuppressive drugs in solid organ transplant recipients.
`MMF is an inhibitor of inosine-5 0 -monophosphate, and is able to
`preferentially inhibit B-cell and T-cell function. The
`immunosuppressive abilities of MMF have made it one of the most
`successful anti-rejection drugs in transplant patients, but patients also
`appear to have increased susceptibility to infections, speci¢cally
`cytomegalovirus and BK virus. Despite its association with an
`increased risk of infection, MMF has also exhibited antimicrobial
`activity against pathogens including hepatitis C, Pneumocystis jirovecii,
`and human immunode¢ciency virus. A thorough understanding of the
`functions of MMF on the immune system and interaction with
`infectious pathogens could be helpful in implementing preventative
`strategies against opportunistic infections in transplant patients.
`
`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`
`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`

`
`M.L. Ritter1, L. Pirofski1,2
`1Department of Medicine, Division of Infectious Diseases,
`Albert Einstein College of Medicine and Montefiore Medical
`Center, Bronx, New York, USA, 2Department of Microbiology
`and Immunology, Albert Einstein College of Medicine, Bronx,
`New York, USA
`
`Key words: mycophenolate mofetil; solid organ transplant;
`cytomegalovirus; BK virus
`
`Correspondence to:
`Liise-Anne Pirofski, MD, Division of Infectious Diseases,
`Albert Einstein College of Medicine, Room 709 Forchheimer
`Bldg., 1300 Morris Park Avenue, Bronx, NY 10461, USA
`E-mail: pirofski@aecom.yu.edu
`
`Received 29 December 2008, revised 10 February and
`4 March 2009, accepted for publication 5 March 2009
`
`DOI: 10.1111/j.1399-3062.2009.00407.x
`Transpl Infect Dis 2009: 11: 290–297
`
`Mycophenolate mofetil (MMF) is a morpholinoethyl ester
`and a prodrug of mycophenolic acid (MPA), a potent inhib-
`itor of inosine-5 0 -monophosphate dehydrogenase (IMPDH)
`(1). F|rst approved by the US Food & Drug Administration
`for use in renal transplantation in 1995, MMF has now been
`used for over 10 years as an immunosuppressive agent in
`solid organ transplant recipients (2). According to the Sci-
`enti¢c Registry of Transplant Recipients, currently 86% of
`renal transplant recipients receive MMF as part of their im-
`munosuppressive regimen, most frequently in combina-
`tions with prednisone (Pr) and tacrolimus (Tac) (3). MMF
`is unique among transplant medications in its ability to in-
`hibit both T-lymphocyte and B-lymphocyte activity.
`Its anti-B-lymphocytic activity has been targeted for the
`treatment of rheumatologic disease, and already has
`proven e⁄cacy against lupus nephritis (4, 5). However,
`the broad immunosuppressive abilities that make MMF
`an e⁄cacious drug for transplant and autoimmune dis-
`eases might also enhance the risk of infectious complica-
`tions (6).
`
`Mechanism of action
`
`The development of MMF as an immunosuppressive agent
`stemmed from observations in patients with 2 genetic ill-
`nesses. Children with an absence of adenosine deaminase,
`an enzyme responsible for the de novo production of guano-
`sine monophosphate, had a combined immunode¢ciency
`involving de¢ciencies in both T and B lymphocytes (7 ).
`However, children born with Lesch^Nyhan syndrome, a de-
`¢ciency of hypoxanthine-guanine phosphoribosyl trans-
`ferase, and thus the salvage production of guanosine mono-
`phosphate, su¡ered from neurologic abnormalities and
`gout, but had normal immune function (8). These 2 obser-
`vations led scientists to look for an inhibitor speci¢c for de
`novo guanosine monophosphate synthesis. MPA, a fermen-
`tation product of Penicillium brevicompactum and related
`fungi, is an inhibitor of IMPDH, and preferentially inhibits
`de novo guanosine synthesis. Depletion of de novo guano-
`sine causes a lack of deoxyguanosine triphosphate, sup-
`
`290
`
`

`

`pressing DNA synthesis and proliferation of T and B lym-
`phocytes (9). An advantage of MPA is its preferential inhi-
`bition of the type II isoform of IMPDH. The type II isoform
`is expressed almost exclusively in activated T and B lym-
`phocytes, while the type I isoform is expressed in most
`other cells. Therefore, the activity of MPA is speci¢cally
`targeted toward T and B lymphocytes (10, 11). MMF is the
`ester prodrug of MPA, and was found to increase oral bio-
`availability of the drug (12). Most studies have looked at the
`function and e¡ects speci¢cally of MPA on immune cells.
`However, because MMF is metabolized in vivo to MPA, no
`di¡erence in immune activity is expected. MMF has now
`become one of the most frequent medications given to pa-
`tients post transplant, and has proved extremely e⁄ca-
`cious in preventing graft rejection.
`
`Activity against T lymphocytes
`
`MPA inhibits T-lymphocyte proliferation by decreasing
`de novo guanosine production and ultimately DNA synthe-
`sis inT lymphocytes. MPA has also been shown to increase
`apoptosis in human T lymphocytes, speci¢cally in acti-
`vated T lymphocytes (9). This was demonstrated by Nak-
`amura et al. (13) when peripheral blood T lymphocytes
`stimulated with staphylococcal enterotoxin B and treated
`with MPA, showed a marked increase in apoptosis.
`Their ¢ndings suggested that MMF can induce apoptosis
`in antigen-activated T lymphocytes (13). Cohn et al. (14)
`also found that MPA increased apoptosis of activated
`T cells, speci¢cally MOLT- 4 cells, a human T-lymphocyte
`cell line derived from a patient with acute lymphoblastic
`leukemia.
`Studies have also examined the e¡ects of MPA on cyto-
`toxic T cells, mostly in an e¡ort to understand the e¡ec-
`tiveness of MPA in preventing transplant rejection. Eugui
`et al. (15) showed that MPA exhibited a dose-dependent de-
`crease in cytotoxic T-cell activity in mice.
`
`Activity against B lymphocytes
`
`As with T lymphocytes, MPA inhibits B lymphocytes via
`inhibition of IMPDH, and thus DNA synthesis in B lym-
`phocytes. In addition, in vitro studies have shown that
`MPA inhibits the proliferation of human B lymphocytes
`and immunoglobulin (Ig) production in response to Staph-
`ylococcus protein A-sepharose (10). Jonsson and Carlsten (16)
`demonstrated that when a B-cell hybridoma was exposed
`to MPA, a decrease in IgG production occurred, as well as
`a decrease in cytokine levels and decreased proliferation of
`the cells.
`
`Ritter & Pirofski: Mycophenolate mofetil
`
`In animal studies, MPA was ¢rst shown to inhibit anti-
`body formation in rats immunized with sheep red blood
`cells (15). Subsequent studies found that MPA inhibited an-
`tibody production in response to in£uenza virus hemag-
`glutinin in mice (9). Kimball et al. (17 ) found that when
`mice treated with equine-derived polyclonal antithymocyte
`immunoglobulin (ATGAM) received MMF, they had a sig-
`ni¢cant decrease in the production of anti-ATGAM anti-
`bodies.
`More recent literature suggests a reduction in pathogen-
`speci¢c Ig production in patients receiving MMF. Zmonar-
`ski et al. (18) studied 33 renal transplant patients who were
`found to have clinical cytomegalovirus (CMV) disease
`while on immunosuppression with 1 of 3 regimens: azathi-
`oprine (Aza) 1 cyclosporine (CyA) 1 Pr, or MMF 1 CyA 1
`Pr, or Tac 1 MMF. The authors found no di¡erence in CMV
`IgG levels between the patients on di¡erent regimens, but
`there was a decrease in CMV IgM production in the pa-
`tients receiving MMF (18).
`
`Other immunosuppressive activity
`
`MPA has also been shown in vivo to inhibit the capacity of
`dendritic cells to e⁄ciently present antigens to T lympho-
`cytes (19). MPA reduces the recruitment of monocytes into
`sites of graft rejection and in£ammation, and also in-
`creases apoptosis of monocytes (9). Studies also have found
`that MPA directly in£uences the function of endothelial
`cells, speci¢cally disrupting leukocyte adhesion. In addi-
`tion, the recruitment of lymphocytes and monocytes into
`in£ammatory tissues is decreased by MPA (9).
`
`Infectious complications associated with
`MMF
`
`Infection in the post-transplantation period is a major
`cause of patient morbidity and is the most frequent cause
`of death of patients in the early post-transplant period (20).
`While the suppressive activity of MPA on multiple immune
`cell types results in decreased rates of transplant rejection,
`which is bene¢cial, it also dampens the normal immune re-
`sponse to infectious agents, which is detrimental. Pour-
`farziani et al. (21) performed a retrospective cohort study
`of patient and graft survival and causes of post-transplant
`admissions in renal transplant recipients who received ei-
`ther Aza or MMF as part of their immunosuppressive regi-
`men. Their results showed that patient and graft survival
`rates were higher in patients who received MMF rather
`than Aza, and that rates of re-hospitalization for rejection
`
`Transplant Infectious Disease 2009: 11: 290^297
`
`291
`
`

`

`Ritter & Pirofski: Mycophenolate mofetil
`
`were lower in MMF recipients. However, the rate of re-hos-
`pitalization for infection was signi¢cantly higher in pa-
`tients who received MMF (50% versus 37%; P 5 0.002),
`though neither the sites of infection nor the speci¢c patho-
`gens involved were described. The authors also found a
`small increase in mortality and intensive care unit admis-
`sions in the MMF cohort (21).
`Another study demonstrated that late introduction of
`MMF, 41 year post transplant, resulted in increased rates
`of infection.Thirty patients receiving a regimen of prednis-
`olone, Cya, and Aza post transplant were switched from
`Aza to MMF. The rate of infection increased signi¢cantly
`in patients after they were switched to MMF, with a pre-
`conversion rate of 26.7% and a post-conversion rate of
`66.6% (Po0.0005). Most post-conversion infections were
`upper respiratory tract infections (Haemophilus in£uenzae
`or Staphylococcus aureus in most cases, with 1 episode of
`Moraxella catarrhalis), followed by urinary tract infections
`(mostly Escherichia coli or Klebsiella species), 1 case of septic
`arthritis with Streptococcus pneumoniae, and 1 case of
`S. pneumoniae septicemia. All infections except 1 occurred
`41 month after the patient was switched to MMF. The rate
`of recurrent infection also increased signi¢cantly post con-
`version to MMF, with a rate of 6.6% before conversion to
`MMF and 43.3% post conversion (Po0.0005). The authors
`suggest that a possible contributing factor was decreased
`renal function post transplant, essentially increasing se-
`rum levels of MMF with standard doses (6).
`
`CMV
`
`The infectious complication most frequently associated
`with MMF is CMV disease. During the initial trials of
`MMF in transplant patients, MMF was associated with an
`increased rate of CMV disease. The US Renal Transplant
`Mycophenolate Mofetil Study Group, in a comparison of pa-
`tients treated with MMF versus Aza, found a dose-depen-
`dent increase in tissue invasive CMV disease with MMF
`versus Aza (10.8% at 3 g/day and 9.1% at 2 g/day versus
`6.1% with Aza), but no statistical analysis was presented
`(22).
`Moreso et al. (23) also found a dose-dependent increased
`rate of CMV disease in patients receiving MMF. In their
`study, patients who received high-dose Pr with Cya and
`MMF at 3 g/day had a very high incidence of CMV dis-
`ease. But if either the Pr or Cya dose was decreased, or if
`the MMF dose was decreased to 2 g/day, the incidence of
`CMV disease decreased (23). However, use of ganciclovir
`prophylaxis in these study populations was not disclosed.
`In 2004,Wang et al. (24) reviewed randomized clinical trials
`in which patients received either Aza or MMF as part of
`
`292
`
`Transplant Infectious Disease 2009: 11: 290^297
`
`their regimen. They looked at a total of 20 trials with 6870
`patients who had undergone renal transplantation. They
`found a statistically signi¢cant increase in CMV disease
`in patients receiving 3 g/day of MMF versus Aza. While
`there was also an increase in CMV disease in patients re-
`ceiving 2 g/day of MMF, the di¡erence was not statistically
`signi¢cant (24). However, several other studies have also
`demonstrated an increased rate of CMV disease in patients
`on MMF at the recommended dose of 2 g/day (25^28). Jorge
`et al. (2) retrospectively examined the outcomes of the 280
`renal transplants performed over a 10 -year period. While
`MMF was successful in reducing early acute rejection and
`prolonging graft survival, there was a signi¢cantly higher
`rate of CMV disease in patients receiving MMF at the stan-
`dard dose of 2 g/day versus Aza (2). A caveat in evaluating
`these studies is that in all 3, the use of ganciclovir prophy-
`laxis was not noted. In both the Moreso and the Wang stud-
`ies (23, 24), MMF was associated with a statistically
`signi¢cant increase in leukopenia, especially at the higher
`dose of 3 g/day. Hence, it remains unclear whether the in-
`crease in CMV disease was secondary to increased leu-
`kopenia, or due to a direct action of MMF itself.
`A recent study by the RESITRA network of the Spanish
`study group of infection in transplantation, which reported
`on the development of CMVdisease after renal transplanta-
`tion, did not list MMF among the risk factors (29). This
`study had the advantage of being very large (1470 renal
`transplant patients) and prospective. CMV disease oc-
`curred in 6.7% of patients, and after univariate and multi-
`variate analyses, the factors independently associated with
`a statistically signi¢cant increase in CMVdisease included
`donor age 460, Cya use, and chronic graft dysfunction.
`However, it is important to note that an analysis of the e¡ect
`of MMFon CMV risk would have been di⁄cult to ascertain,
`because a majority of the study patients (83.7%) received
`MMF as part of their post-transplant immunosuppressive
`regimen. Along the same lines, it is also noteworthy that
`use of sirolimus was associated with a decreased incidence
`of CMV disease, and that there was a statistically signi¢-
`cant decrease in the use of MMF in patients who were tak-
`ing sirolimus. Therefore, although the RESITRA study is
`important for advancing our knowledge of risk factors for
`CMV disease, it does not directly address the question of
`CMV disease as it pertains to MMF use.
`Zmonarski et al. (18) found that patients with CMV
`disease who were receiving MMF had decreased levels of
`serum anti-CMV IgM compared with patients who were re-
`ceiving Aza. In this study, 75 patients who underwent renal
`transplantation were monitored for the presence of CMV
`pp65 antigen. If the patient became pp65 -positive 1^5
`months post transplant, they were enrolled in the study. Se-
`rum CMV IgG and IgM levels were measured at the time of
`transplantation, 1^5 months after transplantation, and at
`
`

`

`the time of diagnosis of CMV disease. While no di¡erence
`was found in CMV IgG levels in patients receiving regi-
`mens containing MMF versus Aza, there were statistically
`signi¢cant lower levels of IgM in the patients receiving reg-
`imens with MMF. Zmonarski et al. (18) suggest that de-
`creased CMV IgM may have important clinical signi-
`¢cance, as it may correlate with more severe CMV disease.
`Unfortunately the study was very small, with a total enroll-
`ment of 33 patients. A retrospective study by Hardwick et
`al. (30) speci¢cally examined CMV IgG levels in renal
`transplant recipients who received either Aza or MMF and
`developed CMV disease. They reviewed serum CMV IgG
`antibody levels as determined by enzyme immunoassay
`(drawn approximately 1 month after transplantation), and
`found no di¡erence between the level of CMV IgG in pa-
`tients on MMF versus Aza (30). As in the Zmonarski study
`(18), the authors did not ¢nd a decrease in CMV IgG in pa-
`tients receiving MMF, but they did not examine CMV IgM
`levels. The underlying vulnerability of patients on MMF to
`CMV disease has yet to be thoroughly understood, but
`these studies raise the possibility that CMV IgM levels
`may correlate with the risk of CMV disease in patients re-
`ceiving MMF.
`Although CMVdisease may occur more often in those pa-
`tients who are on MMF, MMF-treated patients tend to have
`better outcomes with CMV. This is believed to be due to a
`synergistic e¡ect of MMF on ganciclovir (31). In a study by
`Giral et al. (32), patients with CMV disease receiving MMF
`had less CMV disease-associated graft loss than patients
`receiving Aza. Hence, although the incidence of CMV dis-
`ease may be higher in patients with MMF, once ganciclovir
`is started, patient outcomes tend to improve. Future studies
`may consider investigating at what point to start gan-
`ciclovir therapy in patients receiving MMF and whether
`preemptive or prophylactic ganciclovir may be safer in this
`more vulnerable population.
`
`Ritter & Pirofski: Mycophenolate mofetil
`
`patients and found that of the patients with BKVN, 6 of
`the 7 were on highly immunosuppressive regimens that in-
`cluded Tac and/or MMF. Shi et al. (38) studied 7 cases of
`BKVN in 80 renal transplant recipients and found that a
`signi¢cantly higher number of patients who developed
`BKVN had been on Tac and MMF. Upon histological exam-
`ination, all of the patients who had severe BKVN had been
`receiving Tac and MMF. In addition, all graft failures oc-
`curred in patients on MMF and Tac (38). F|nally, Barton et
`al. (39) found that MMF use was an independent risk factor
`for development of BK viruria in non-renal solid organ
`transplant recipients. In 34 non-renal solid organ trans-
`plant (lung, liver, heart, heart-lung) patients with chronic
`renal dysfunction, 5 had BKV viruria, all of whom were re-
`ceiving MMF (5 of 19 [26%] versus 0 of 15 who were not;
`P 5 0.03) (39).
`While agents such as cidofovir and le£unomide have
`been investigated as possible therapies for BKVN, at this
`time the standard treatment is reduction in or removal of
`immunosuppression, which again emphasizes the essen-
`tial role that immunosuppressive drugs play in the develop-
`ment of BKVN (40).
`
`Other infections associated with MMF
`
`MMF use has also been associated with increased suscep-
`tibility to varicella disease. Lauzurica et al. (40) published 4
`case reports of disseminated varicella among 275 renal
`transplant recipients. The authors noted that, before the
`initiation of MMF, they had never seen a case of varicella
`in their transplant recipients. However, other studies have
`not found a signi¢cant association (41).
`Meier-Kriesche et al. (27 ) found a statistically signi¢cant
`increase in fungal infections in their geriatric renal trans-
`plant population receiving MMF versus Aza, but the spe-
`ci¢c organisms and sites of infection were not reported.
`
`BKV
`
`BKV infection has emerged as an especially important in-
`fection in the transplant recipient. BK virus nephropathy
`(BKVN) has been associated with allograft dysfunction
`and graft failure in 1^8% of renal transplant recipients,
`with a loss of the allograft occurring in half the cases (33^
`35).
`BKV viremia has been associated with the use of higher
`dose immunosuppressive regimens (36). Several studies
`have shown that regimens that include MMF with Tac in-
`crease the incidence of BKV viremia. Mengel et al. (37 ) ex-
`amined 1276 renal biopsies from 638 renal transplant
`
`Possible antimicrobial activity of MMF
`
`Interestingly, while MMF is associated with an increased
`risk of infection, published reports suggest that it may have
`antimicrobial e¡ects against certain pathogens (Table 1)
`(42^52). The ¢rst published reports of the antimicrobial
`properties of mycophenolate were published in the 1960s
`when in vitro studies showed inhibition of vaccinia virus,
`herpes simplex virus, Coxsackie virus, and in£uenza virus
`by MMF (53, 54). Since that time, studies have identi¢ed
`other infections inhibited by MMF.
`
`Transplant Infectious Disease 2009: 11: 290^297
`
`293
`
`

`

`Ritter & Pirofski: Mycophenolate mofetil
`
`The e¡ects of mycophenolate mofetil (MMF) on select infections
`
`Related studies
`
`Pathogen
`
`Author (year), reference
`
`Findings
`
`Proposed mechanism of action
`
`Dengue virus
`
`Diamond et al. (2002 ),
`
`In vitro, MPA blocked DV infection of hepatoma cell lines
`
`MPA prevents the synthesis and
`
`(DV)
`
`(42 )
`
`by 99%
`
`accumulation of DV RNA
`
`Takhampunya et al.
`
`(2006 ), (43 )
`
`In vitro, MPA resulted in a ¢vefold increase in defective DV
`RNA production in monkey kidney cells
`
`MPA depletes intracellular GTP pool,
`
`inhibiting DV RNA production and viral
`
`replication
`
`Hepatitis C
`
`virus (HCV)
`
`Hepatitis B
`
`virus (HBV)
`
`Henry et al. (2006), (44)
`
`In vitro HCV-replication model with a luciferase reporter
`
`Unknown, but appears to be independent
`
`Jain et al. (2002 ), (45)
`
`Randomized clinical trial of patients with HCV undergoing
`
`MMF either has no anti-HCV activity, or its
`
`gener; MPA inhibited HCV replication to 75%; Inhibition
`
`of guanosine depletion
`
`was independent of guanosine production
`
`liver transplant; received either TAC plus prednisone, or
`
`immunosuppressive properties
`
`TAC plus prednisone plus MMF; MMF had no impact on
`
`overwhelm its antiviral e¡ect in the
`
`patients survival, graft survival, rejection, or rate of HCV
`
`clinical setting
`
`recurrence
`
`Wu et al. (2003 ), (46)
`
`In vitro, 2.2.15 cells were treated with di¡erent
`
`MPA suppresses expression of HBsAg
`
`concentrations of MPA. MPA decreased expression of
`
`and HBeAg as well as replication of HBV
`
`HBsAg, HBeAg, and HBV DNA in a dose-dependent
`
`DNA
`
`manner
`
`Ben Ari et al. (2001), (47)
`
`Prospective study of 4 liver allograft recipients with
`
`MMF at the dosage used is not e¡ective
`
`recurrent HBV infection resistant to lamivudine received
`
`at suppressing HBV replication
`
`MMF, in addition to decreased doses of corticosteroid
`
`and cyclosporine. After 8 weeks of therapy, serum ALT
`
`increased in 2 patients, remained stable in 1 patient, and
`
`decreased in 1. Serum HBV DNA levels increased in 3
`
`Pneumocystis
`
`Oz et al. (1997), (48)
`
`Virus-free Sprague^Dawley rats were treated with TAC,
`
`Unclear mechanism by which MMF
`
`jirovecii
`
`sirolimus, dexamethasone, and/or MMF. Increased rates
`
`appears to have antimicrobial action
`
`patients, and decreased in 1
`
`of PCP seen in rats which received TAC, sirolimus, or
`
`against Pneumocystis
`
`dexamethasone, but none of the rats treated with MMF
`
`or MMF plus dexamethasone had PCP
`
`Husain et al. (2002 ), (49)
`
`Combined data from 4 clinical trials where patients
`
`Unclear mechanism by which MMF exerts
`
`received MMF. None of the patients receiving MMF
`
`protection against PCP
`
`developed PCP (0/1068) while 10 of the patients not
`
`receiving MMF developed PCP (1.8%)
`
`Coxsackie
`
`Padalko et al. (2003 ),
`
`C3H-mice were infected with CBV3 and received twice
`
`Unclear, but not believed to be due to
`
`virus B3
`
`(CVB3 )
`
`(50)
`
`daily MMF for 7 days. Mice who received MMF had
`
`inhibition of viral replication since the
`
`signi¢cant reduction in the development of CBV-induced
`
`titers of infectious virus and viral RNA
`
`myocarditis (dose dependent)
`
`were increased in mice treated with MMF
`
`West Nile virus
`
`Morrey et al. (2002 ), (51)
`
`MA-104 cells infected with NewYorkWNV; assays used to
`
`measure inhibition of virus showed MPA to be one of the
`
`most potent antivirals; it was even more active against a
`
`Ugandan strain of WNV
`
`Unclear antiviral activity in vitro against
`WNV
`
`(WNV)
`
`Yellow fever
`
`virus (YFV)
`
`Neyts et al. (1996 ), (52 )
`
`Used the attenuated vaccine strain 17D of YFV to
`
`MPA has anti-YFV activity, but it is not
`
`evaluate inhibitory e¡ect in infected Vero cells. MPA was a
`
`merely cytotoxic, given the lack of activity
`
`strong inhibitor of YFV, but had little or no e¡ect on Bunya
`
`against Bunya virus PuntaToro and HSV
`
`virus PuntaToro or HSV
`
`GTP, guanosine triphosphate; TAC, tacrolimus; MPA, mycophenolic acid; HBsAg, hepatitis B surface antigen; HBeAg, hepatitis B virus early antigen;
`
`ALT, alanine aminotransferase; PCP, Pneumocystis jirovecii pneumonia; TAC, tacrolimus; HSV, herpes simplex virus.
`
`Table1
`
`In vitro studies with dengue virus showed that MPA com-
`pletely inhibited infection of human cells by the virus (42,
`43). In patients with chronic hepatitis C virus (HCV) dis-
`ease, MMF was shown to be a potent in vitro inhibitor of
`
`HCV replication. Interestingly, the inhibition was indepen-
`dent of cell proliferation and guanosine depletion, so the
`true mechanism of action is unknown (44). However, a pro-
`spective randomized trial of MMF in liver transplants with
`
`294
`
`Transplant Infectious Disease 2009: 11: 290^297
`
`

`

`HCV showed no impact on patient survival, graft survival,
`rejection, or rate of HCV recurrence. The authors suggest
`that while MMF has in vitro activity against HCV, the over-
`all immunosuppressive properties of the drug may domi-
`nate in vivo (45). MMF was also found to have some
`activity against hepatitis B virus in vitro (46), but has not
`been found to be e¡ective in vivo (47 ).
`MMF has been shown to have a possible protective e¡ect
`against Pneumocystis jirovecii pneumonia (PCP) (48, 49).
`This has raised the question of the necessity of PCP pro-
`phylaxis in patients on MMF. Isolated reports suggest
`MMF may also have activity against Coxsackie virus (50),
`West Nile virus (51), and yellow fever virus (52). At this
`point, MMF is not a standard therapy for any of these infec-
`tions.
`F|nally, MMF has demonstrated activity against human
`immunode¢ciency virus (HIV) both in vivo and in vitro (55^
`61). Its role against HIV is somewhat controversial. MPA is
`consistently shown to increase the activity of other antiret-
`rovirals, but it is unclear if MPA alone has e¡ective activ-
`ity against the virus (62, 63). MPA is known to be active
`against HIV especially in combination with abacavir (64,
`65). The mechanism of anti-HIV activity is believed to
`be secondary to guanosine triphosphate depletion, with
`the inhibition of reverse transcriptase, and also through a
`reduction in gp120 expression in transformed cells (66).
`Several studies suggested that MPA may have a role in the
`treatment of multidrug-resistant HIV infection; however,
`clinical trials have failed to show a signi¢cant bene¢t from
`the addition of MMF to anti-retroviral regimens. Sankat-
`sing et al. (55) found that MMF did not signi¢cantly in-
`crease either the plasma HIV-1 RNA decay rate or the
`decay rate of latently infected cells when it was added to a
`triple class antiretroviral regimen in treatment-na|« ve pa-
`tients.
`
`Conclusion
`
`As the use of MMF increases in transplant recipients and
`in autoimmune diseases, it has become more important to
`fully understand its impact on infectious complications.
`Further studies are needed to investigate how the e¡ects
`of MMF on immune cells predispose patients to speci¢c in-
`fections. Such information may make it possible to antici-
`pate the infections, and help determine the choice and
`length of prophylactic antimicrobials in transplant recipi-
`ents receiving MMF.
`In addition, the possible antimicrobial activities of MMF
`could be utilized in sparing patients from unnecessary pro-
`phylaxis, or in guiding the use of MMF in speci¢c popula-
`
`Ritter & Pirofski: Mycophenolate mofetil
`
`tions. For instance, if further studies are able to fully
`decipher the relationship between MMF and PCP, an argu-
`ment could be made for modifying PCP prophylaxis in pa-
`tients receiving MMF. In doing such, patients could be
`spared the possible hematopoietic side e¡ects of trime-
`thoprim-sulfamethoxazole. Given the in vivo activity of
`MMF against HIV, future studies could investigate the pos-
`sible bene¢ts of using MMF in HIV-positive transplant pa-
`tients.
`A better understanding of the activity of MMF against
`these infections could bene¢t future transplant recipients
`and improve the overall outcomes of patients in the post-
`transplant period.
`
`Acknowledgement:
`
`This work was supported by grants from National Insti-
`tutes of Health Grants R01AI045459 and R01AI035370
`to L.P.
`
`References
`
`1. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanism of
`action. Immunopharmacology 2000; 47: 85^118.
`2. Jorge S, Guerra J, Santana A, Mil-Homens C, Prata MM.
`Mycophenolate mofetil: ten years’ experience of a renal transplant
`unit. Transplant Proc 2008; 40: 700^704.
`3. OPTN/SRTR Annual Report. Immunosuppression use for
`maintenance by regimen prior to discharge, 1997-2006 Recipients
`with kidney transplants. 2007. Available at http://
`www.ustransplant.org (accessed September 9, 2008).
`4. Lu F,Tu Y, Peng X, et al. A prospective multicentre study of
`mycophenolate mofetil combined with prednisolone as induction
`therapy in 213 patients with active lupus nephritis. Lupus 2008; 17:
`622^629.
`5. Bao H, Liu ZH, Xie HL, Hu WX, Zhang HT, Li LS. Successful
`treatment of class V 1 IV lupus nephritis with multitarget therapy. J
`Am Soc Nephrol 2008; 19: 2001^2010.
`6. Hanvesakul R, Kubal C, Jham S, et al. Increased incidence of
`infections following the late introduction of mycophenolate mofetil in
`renal transplant recipients. Nephrol Dial Transplant 2008; 23 (12):
`4049^4053.
`7. Giblett ER, Anderson JE, Cohen F, Pollara B, Meuwissen HJ.
`Adenosine-deaminase de¢ciency in two patients with severely
`impaired cellular immunity. Lancet 1972; 2: 1067^1069.
`8. Allison AC, Hovi T,Watts RW,Webster AD. Immunological
`observations on patients with Lesch^Nyhan syndrome, and on the
`role of de-novo purine synthesis in lymphocyte transformation.
`Lancet 1975; 2: 1179^1183.
`9. Allison AC, Eugui EM. Mechanisms of action of mycophenolate
`mofetil in preventing acute and chronic allograft rejection.
`Transplantation 2005; 80: S181^S190.
`10. Eugui EM, Almquist SJ, Muller CD, Allison AC. Lymphocyte-selective
`cytostatic and immunosuppressive e¡ects of mycophenolic acid in
`vitro: role of deoxyguanosine nucleotide depletion. Scand J Immunol
`1991; 33: 161^173.
`
`Transplant Infectious Disease 2009: 11: 290^297
`
`295
`
`

`

`Ritter & Pirofski: Mycophenolate mofetil
`
`11. Carr SF, Papp E,Wu JC, NatsumedaY. Characterization of human type
`I and type II IMP dehydrogenases. J Biol Chem 1993; 268: 27286^
`27290.
`12. Allison AC, Almquist SJ, Muller CD, Eugui EM. In vitro
`immunosuppressive e¡ects of mycophenolic acid and an ester pro-
`drug, RS- 61443. Transplant Proc 1991; 23: 10^14.
`13. Nakamura M, Ogawa N, Shalabi A, Maley WR, Longo D, Burdick JF.
`Positive e¡ect on T-cell regulatory apoptosis by mycophenolate
`mofetil. Clin Transplant 2001; 15 (Suppl. 6): 36^40.
`14. Cohn RG, Mirkovich A, Dunlap B, et al. Mycophenolic acid increases
`apoptosis, lysosomes and lipid droplets in human lymphoid and
`monocytic cell lines. Transplantation 1999; 68: 411^418.
`15. Eugui EM, Mirkovich A, Allison AC. Lymphocyte-selective
`antiproliferative and immunosuppressive e¡ects of mycophenolic
`acid in mice. Scand J Immunol 1991; 33: 175^183.
`16. Jonsson CA, Carlsten H. Mycophenolic acid inhibits inosine 5 0 -
`monophosphate dehydrogenase and suppresses immunoglobulin and
`cytokine production of B cells. Int Immunopharmacol 2003; 3: 31^37.
`17. Kimball JA, Pescovitz MD, Book BK, Norman DJ. Reduced human IgG
`anti-ATGAM antibody formation in renal transplant recipients
`receiving mycophenolate mofetil. Transplantation 1995; 60: 1379^
`1383.
`18. Zmonarski SC, Boratynska M, Madziarska K, et al. Mycophenolate
`mofetil severely depresses antibody response to CMV infection in
`early posttransplant period. Transplant Proc 2003; 35: 2205^2206.
`19. Mehling A, Grabbe S,Voskort M, Schwarz T, Luger TA, Beissert S.
`Mycophenolate mofetil impairs the maturation and function of
`murine dendritic cells. J Immunol 2000; 165: 2374^2381.
`20. Pourmand G, Salem S, Mehrsai A,Taherimahmoudi M, Ebrahimi R,
`Pourmand MR. Infectious complications after kidney
`transplantation: a single-center experience. Transpl Infect Dis 2007;
`9: 302^309.
`21. Pourfarziani V, Panahi Y, Assari S, Moghani-Lankarani M, Saadat SH.
`Changing treatment protocol from azathioprine to mycophenolate
`mofetil: decrease in renal dysfunction, increase in infections.
`Transplant Proc 2007; 39: 1237^1240.
`22. Sollinger HW. Mycophenolate mofetil for the prevention of acute
`rejection in primary cadaveric renal allograft recipients. U.S. Renal
`Transplant Mycophenolate Mofetil Study Group. Transplantation
`1995; 60: 225^232.
`23. Moreso F, Seron D, Morales JM, et al. Incidence of leukopenia and
`cytomegalovirus disease in kidney transplants treated with
`mycophenolate mofetil combined with low cyclosporine and steroid
`doses. Clin Transplant 1998; 12: 198^205.
`24. Wang K, Zhang H, Li Y, et al. Safety of mycophenolate mofetil versus
`azathioprine in renal transplantation: a systemic review. Transplant
`Proc 2004; 36: 2068^2070.
`25. Basic-Jukic N, Kes P, Bubic

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket