Skip to main content

Main menu

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Podcasts
    • Subject Collections
    • Archives
    • Kidney Week Abstracts
    • Saved Searches
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Trainees
    • Peer Review Program
    • Prize Competition
  • About CJASN
    • About CJASN
    • Editorial Team
    • CJASN Impact
    • CJASN Recognitions
  • More
    • Alerts
    • Advertising
    • Feedback
    • Reprint Information
    • Subscriptions
  • ASN Kidney News
  • Other
    • ASN Publications
    • JASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology

User menu

  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
American Society of Nephrology
  • Other
    • ASN Publications
    • JASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Advertisement
American Society of Nephrology

Advanced Search

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Podcasts
    • Subject Collections
    • Archives
    • Kidney Week Abstracts
    • Saved Searches
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Trainees
    • Peer Review Program
    • Prize Competition
  • About CJASN
    • About CJASN
    • Editorial Team
    • CJASN Impact
    • CJASN Recognitions
  • More
    • Alerts
    • Advertising
    • Feedback
    • Reprint Information
    • Subscriptions
  • ASN Kidney News
  • Visit ASN on Facebook
  • Follow CJASN on Twitter
  • CJASN RSS
  • Community Forum
Moving Points in Nephrology
You have accessRestricted Access

Presensitization: The Problem and Its Management

Stanley C. Jordan and Mark D. Pescovitz
CJASN May 2006, 1 (3) 421-432; DOI: https://doi.org/10.2215/CJN.01651105
Stanley C. Jordan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mark D. Pescovitz
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data Supps
  • Info & Metrics
  • View PDF
Loading

Abstract

Much attention has been placed recently on transplantation in highly HLA-sensitized patients. In attempts to remove these antibodies and enable successful transplantation, several novel approaches have been developed. These include intravenous Ig (IVIg), mycophenolate mofetil, sirolimus, alemtuzumab, protein A immunoabsorption, and rituximab. IVIg has emerged as a very effective agent when used alone in high dose or when used in low dose and combined with plasmapheresis. Although alemtuzumab has been used to eliminated B cells, it fails to prevent antibody-mediated rejection and therefore probably is not suitable for desensitization. Rituximab, a B cell-specific antibody, seems to be safe and to have some efficacy as a sole agent in elimination of alloantibodies but most likely will require combination therapy with IVIg or other agents. Newer agents, such as humanized anti-CD20, are being developed. Despite the great interest in the problem of allosensitization, with one notable exception, there is a major deficiency in controlled clinical trials, the conduct of which should be a focus for the near future.

In patients with high levels of preformed anti-HLA antibodies (high panel-reactive antibodies [PRA]; highly sensitized), transplant rates are dramatically reduced because of the additional immunologic barrier with increased rejection risk. From 1994 to 2003, the numbers of highly sensitized patients who were on the transplant waiting list continued to increase (12,808 in 1994 versus 17,814 in 2003) (1). In 2003, 32% of the transplant waiting list was considered sensitized to HLA antigens, with 13.7% having PRA >80% (1). These antibodies result from exposure to non-self HLA antigens, usually from previous transplants, blood transfusions, and/or pregnancies (2).

In 2003, only 6.5% of all kidney transplants that were performed in the United States were in patients with PRA >80%, despite representing approximately 14% of the waiting list (1,3). When these patients receive a transplant, they experience an increased number of rejection episodes and have poorer graft survival (4). The highly sensitized patient is destined to remain on the waiting list for extended periods on dialysis, an added risk factor for decreased patient and graft survival (1,3,5–7). The financial and emotional costs of maintaining highly sensitized transplant candidates on dialysis for years are considerable and contrast greatly with the benefits that are provided by a successful transplant. Therefore, early transplantation results in considerable cost savings, reduced morbidity and mortality, and improvement in quality of life, an elusive goal until recently.

Patel and Terasaki (4) demonstrated that poor outcomes resulted when kidneys were transplanted across a positive cross-match (CMX) barrier and established the basis for modern CMX testing as a means of allocating kidneys. Sensitization is a significant barrier to successful kidney transplantation.

The risks for transplantation can be assessed using currently available standard assays. Today, the techniques that are used to detect anti-HLA antibody include cytotoxicity (CDC) with/without anti-human globulin, ELISA, and flow cytometry (using cells and antigen-coated beads). The development of newer, more sensitive assays has led to an increased ability to define highly sensitized patients and identify donor-specific antibody in patients with antibody-mediated rejection (AMR).

The presence of IgG complement-fixing antibody that is specific for donor HLA antigen (class I or class II) without the addition of anti-human globulin represents an unequivocal contraindication to transplantation. Patients who receive a transplant across this barrier are at a very high risk for AMR. The risk is considered moderate to high when antibody detection requires the use of an anti-globulin reagent in the cytotoxicity assay or the use of a binding assay (e.g., ELISA, flow beads). Other factors, such as history of sensitizing events and titer and duration of anti-HLA antibody, also are important risk factors for AMR.

Until recently, no therapeutic approaches existed to deal with this problem. Currently, two primary protocols have emerged. These include the plasmapheresis/cytomegalovirus Ig (CMV-Ig) protocol (Johns Hopkins Protocol) (8) and the high-dose intravenous Ig (IVIg) protocol (Cedars-Sinai Protocol) (5,9–12). The Mayo Clinic (13) also has extensive experience with both protocols. Both protocols have met with significant success and are discussed here. In addition, the use of other techniques, such as B cell elimination with alemtuzumab, newer immunosuppressive agents, protein A absorption, plasma exchange with concomitant Prograf (Ascellas Pharmaceuticals, Deerfield, IL) and Cellcept (Roche Inc., Nutley, NJ) therapy, and rituximab, are being investigated. These approaches also are discussed here.

Mechanism of Action of IVIg

IVIg products are known to have powerful immunomodulatory effects on inflammatory and autoimmune disorders (14). IVIg products are derived from the plasma of thousands of donors, thereby ensuring a wide diversity of antibody repertoire. Numerous proposed mechanisms of action may be relevant to the modification of allosensitization: (1) Modification of autoantibody and alloantibody levels through induction of anti-idiotypic circuits (5,9–12,14), (2) inhibition of cytokine gene activation and anticytokine activity (14), (3) anti-T cell receptor activity (14), (4) Fc receptor-mediated interactions with antigen-presenting cells (APC) to block T cell activation (14–16), (5) anti-CD4 activity (15), (6) stimulation of cytokine receptor antagonists (14), and (7) inhibition of complement activity (14,17,18). Using the mixed lymphocyte culture system, we have shown that IVIg can significantly inhibit T cell activation and reduce the expression of CD40, CD19, intracellular adhesion molecule-1, CD86, and MHC class II on APC in the mixed lymphocyte reaction (15). The primary effect is on B cells, and, indeed, we have demonstrated that IVIg induces significant B cell apoptosis in vitro through Fc receptor-dependent mechanisms (15). Samuelsson et al. (19) recently described another unique immunoregulatory effector function for IVIg. These investigators demonstrated that IVIg induces the expression of FcγRIIB, an inhibitory receptor on B cells. Therefore, IVIg may have beneficial effects in inflammatory disorders by decreasing B cell activation through interactions with FcγIIB (19). Another interesting observation that may have relevance, especially for the treatment of AMR, is from Magee et al. (17), who showed that IVIg treatment significantly prolonged the survival of pig-to-baboon xenotransplants (from 30 to 60 min to 10 d). This beneficial effect was through inhibition of complement-mediated endothelial cell injury by IVIg. The Fc portion of IVIg has high affinity for activated complement components (C3b and C4b) and could represent a novel mechanism for inhibition of complement-mediated injury to allografts that was described recently for both acute rejection and chronic rejection in humans (18,20,21). Other investigators showed recently that IVIg inhibits the generation of C5b-C9 membrane attack complex, thereby preventing AMR. IVIg also inactivates C3b and accelerates C3b catabolism (14,18). IVIg can inhibit the activation of endothelial cells in in vitro models of inflammation. These observations may have relevance to acceptance of human solid-organ transplants since Williams et al. (21) showed recently that a critical difference between xenografts that survived through accommodation versus those that were lost by AMR was the lack of C5b-C9 membrane attack complex in the grafts with accommodation. Data by Bayry et al. (16) suggest that IVIg inhibits the maturation and function of dendritic cells, impairing their APC activity and inducing IL-10 production. These data are in concert with data from our laboratory demonstrating similar effects on B cells (15). Recently, Abe et al. (22) examined gene expression in patients with Kawasaki disease before and after high-dose IVIg infusion. These investigators demonstrated that in Kawasaki disease, the immunomodulatory effects of IVIg likely were mediated by suppression of an array of immune activation genes in monocytes and macrophages. Gill et al. (23), using an animal model system of ischemia-reperfusion injury, showed that IVIg has direct inhibitory effects on leukocyte recruitment in vitro and in vivo through inhibition of selectin and integrin functions.

Clinical Use of IVIg in Kidney Transplantation

At Cedars-Sinai, we have developed a method to predict which patients would benefit most from IVIg as a desensitization tool. This is summarized in Figures 1 and 2. Figure 1A shows the traditional approach to the highly sensitized patient who has a positive CMX with a living donor. This patient is shifted to the waiting list with monthly antibody screens. The wait times for these patients are exceedingly long (5 to 10 yr) until a suitable CMX(−) kidney becomes available. From our standpoint, this is an unacceptable approach.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

(A) The traditional approach to the highly HLA-sensitized patient. Briefly, for living-donor recipients, if the cross-match (CMX) is negative by CDC and flow, then a transplant is performed. However, if the CMX is positive, the patient is referred to the waiting list until a negative CMX kidney becomes available. This may take >5 yr. In addition, the costs of antibody screening (A/S) and quarterly panel-reactive antibodies (PRA; Q/S) as well as repeat CMX tests with deceased donors is required. The same is true for highly HLA-sensitized patients who are awaiting a deceased-donor kidney. Wait times are extensive (>5 yr), and extensive antibody testing is required. (B) How intravenous Ig (IVIg) in vitro testing for both living-donor recipients and those awaiting a deceased-donor transplant with sufficient wait time can enhance transplantability. Briefly, if the IVIg in vitro test shows any inhibition of the CDC CMX test, then the highly HLA-sensitized patients are treated with IVIg monthly ×4 until a negative or acceptable CMX (living donor) or deceased-donor transplant becomes available. Modification of antibody screens and antibody specificity by IVIg treatment can be monitored as usual and used to guide future therapy. If no inhibition is seen, then the work-up would stop or patients would be referred for plasma exchange therapy (Figure 2).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Approach to highly HLA-sensitized patients who fail in vitro IVIg inhibition or who have very high-titer anti-HLA antibody. The protocol included five plasma exchange treatments followed by IVIg 2 g/kg ×1 and Rituxan 375 mg/m2 ×1. Patients who are entered in this protocol usually are awaiting living-donor transplantation; however, we also would consider this approach for the highly HLA-sensitized patient who is awaiting a deceased-donor transplant if the patient has been on the list for an extensive time (>5 yr) and has frequent offers of deceased-donor organs (dotted line). In addition, patients who are not suitable for IVIg desensitization may be referred to donor exchange programs before or after undergoing plasma exchange therapy. Offering different options will help achieve transplantation for more patients.

In the early 1990s, we developed the IVIg-PRA/CMX test to determine whether IVIg could inhibit PRA or CMX positivity of patients’ sera. Although alternative explanations for the in vitro inhibitory effects of IVIg on cytotoxicity in the PRA and CMX systems have emerged (9,24), we believe that this test is helpful in predicting which patients are likely to benefit from IVIg therapy. This has led to the development of a new paradigm (Figure 1B) in which in vitro reductions of PRA or CMX allow the patients to be treated with high-dose IVIg to enhance their chances for a successful transplant. Transplantation is performed when a negative or acceptable CMX is achieved. In our program, an acceptable CMX is defined as a negative CDC CMX with flow CMX positivity <200 channel shifts for T cells and B cells. Patients who undergo desensitization often have positive flow cytometry CMX at the time of transplantation. Others have described this as well (25).

Figure 2 shows that alternative approaches to desensitization also are necessary. These include plasmapheresis plus IVIg and potential donor exchange programs that have been developed recently at Johns Hopkins. Patients can progress through the various treatment options until successful transplantation is achieved.

IVIg therapy that is given to highly sensitized patients results in reduced allosensitization, reduced ischemia-reperfusion injuries, fewer acute rejection episodes, and higher successful long-term allograft outcomes for cardiac and renal allograft recipients (5,9–12,25–29). Pretreatment with IVIg results in reductions of anti-HLA antibodies and is effective in treatment of allograft rejection episodes (10,28,29). The high-dose IVIg protocol that was developed at Cedars-Sinai evolved from reported efficacy with other inflammatory disorders (e.g., Kawasaki disease) (14). Using the high-dose IVIg protocol (2 g/kg) for desensitization requires that antibody specificity be determined. When IVIg shows any reduction of T or B cell cytotoxicity, using the in vitro assay described above, we treat the recipient with 2 g/kg IVIg (maximum dose 140 g) monthly until the CMX is negative or acceptable, typically with no more than four doses. We also have adapted this to use for highly sensitized deceased-donor transplant candidates who have been on the United Network for Organ Sharing list for >5 yr, have a PRA of >50%, and receive frequent offers for kidneys from donors with whom they have a positive CMX. These patients have an in vitro IVIg PRA, and if suppression or inhibition of the PRA is seen with IVIg, the patients are offered IVIg 2 g/kg monthly ×4 in hopes of achieving desensitization and receiving a CMX-compatible kidney or other organ (Figure 3).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

(A) Summary of the IVIg desensitization protocol for patients with living donors. If IVIg shows in vitro inhibition or reduction of the donor-specific CMX, then IVIg is given at 2 g/kg (maximum dose 140 g) up to monthly ×4 until a negative CMX is achieved. When a negative or acceptable CMX is achieved, the patient is scheduled for transplantation. The patient receives an additional 2 g/kg at 1 mo after transplantation. (B) Protocol for patients who are awaiting deceased donor transplantation. Briefly, a similar protocol for living donors is used. If IVIg shows in vitro reduction of PRA, then patients receive 2 g/kg IVIg monthly ×4. Sera are sent to the Organ Procurement Organization after each infusion and cross-matched with potential deceased donors.

For patients who do not show reductions in PRA or CMX activity in vitro or those who have very high-titer antibodies and are poorly responsive to IVIg, we have developed a modified plasmapheresis/IVIg protocol. This differs from the Johns Hopkins protocol in that only five plasmapheresis treatments are given. This is followed by IVIg 2 g/kg intravenously and Rituxan 375 mg/m2× 1. With this approach, patients have received successful transplants. To date, none has had acute rejection episodes (Figure 4).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

For patients who show no in vitro inhibition of CDC CMX or PRA, we would defer to a plasma exchange protocol shown here. Briefly, the patient receives five plasma exchange treatments over 3 wk. After the last treatment, a repeat CMX with the donor is performed and the patient receives IVIg 2 g/kg ×1. The next day, the patient receives Rituxan 375 mg/m2× 1, and if the CMX is negative or acceptable T cell (flow CMX <200 channel shifts), the transplant is performed.

From July 2002 to October 2005, we evaluated 89 patients who were highly HLA sensitized and had positive CMX with potential donors in the in vitro IVIg-PRA test system. Eighty-five percent showed inhibition to some degree in the in vitro PRA or CMX system. Seventy-nine (89%) of eighty-nine received a transplant after IVIg desensitization therapy (46 living donor, 33 deceased donor). Of the 10 patients who did not receive a transplant, six are awaiting a cadaver transplant offer and two did not respond to IVIg. Two others were successfully desensitized for living donors, but medical conditions prevented transplantation. Therefore, only two (2.2%) of 89 failed to respond to IVIg sufficiently to allow transplantation to be considered. The mean PRA for the cadaver recipients were 83%, and nearly all patients had antibodies specific to their donors that were eliminated or reduced by IVIg therapy. The incidence of allograft rejection is 28% with a 3-yr patient and graft survival of 97.5 and 87.1%, respectively. Five grafts were lost to rejection. The mean serum creatinine at 3 yr was 1.4 mg/dl.

National Institutes of Health IGO2 Study

From 1997 to 2000, the National Institutes of Health (NIH) conducted the IGO2 study that was a controlled, clinical, multicenter, double-blinded trial of IVIg versus placebo in highly sensitized patients who were awaiting kidney transplantation. The study was designed to determine whether IVIg could reduce PRA levels and improve rates of transplantation without concomitantly increasing the risk for graft loss in this difficult-to-transplant group. This study represents the only controlled clinical trial of a desensitization therapy (5). IVIg was superior to placebo in reducing anti-HLA antibody levels (P = 0.004, IVIG versus placebo) and improving rates of transplantation. The 3-yr follow-up showed that the predicted mean time to transplantation was 4.8 yr in the IVIg group versus 10.3 yr in the placebo group (P = 0.02). With a median follow-up of 2 yr after transplantation, the viable transplants functioned normally with a mean (±SE) serum creatinine of 1.68 ± 0.28 (IVIg) versus 1.28 ± 0.13 mg/dl for placebo (P = 0.29). Allograft survival also was superior in the IVIg group at 3 yr. From this multicenter, double-blinded, placebo-controlled trial, we concluded that IVIg is superior to placebo in reducing anti-HLA antibody levels and improving transplantation rates in highly sensitized patients with ESRD. Although more acute rejection episodes were seen in the IVIg treatment group, the 3-yr allograft survival and allograft function were similar to the placebo group. Transplant rates for highly sensitized patients who had ESRD and were awaiting kidney transplants were improved with IVIg therapy. Therefore, IVIg alone offers significant benefits in desensitizing highly HLA-sensitized patients and improves the rates of transplantation in this difficult-to-transplant group without patients’ experiencing excessive allograft loss.

Plasmapheresis/CMV-Ig Protocol

In 1998, Johns Hopkins University Hospital (JHH) began using an intensive preconditioning protocol to allow transplantation across a (+)CMX barrier. Plasmapheresis + CMV-IgG, (PP/CMV-Ig) and a cocktail of immunosuppressive agents are initiated before renal transplantation (30). After transplantation, additional treatments are delivered during the first 10 d (Table 1). The end point of therapy is the elimination of anti-HLA donor-specific antibody either before or after the transplant. The treatment plan is individualized on the basis of an assessment of the patient’s risk of AMR (30). This group identified recipient features that were associated with increased risk for AMR and graft loss. Patients who were thought to be at low risk (e.g., first transplant with pregnancy as sensitizing event) were treated with PP/CMV-Ig and quadruple sequential immunosuppression, whereas high-risk patients (e.g., third transplant with multiple repeat mismatches) have splenectomy and/or anti-CD20 (rituximab, see below) added to their basic treatment plan.

View this table:
  • View inline
  • View popup
Table 1.

Preconditioning regimen for recipients of a (+)CMX or ABO-incompatible renal allograft (JHH PP + CMV-Ig protocol)a

This protocol produces a rapid reduction in anti-HLA titers that allows for transplantation after four to five plasmapheresis treatments. The JHH group members believe that the addition of CMV-Ig adds an immunomodulatory effector mechanism that benefits in keeping the antibody titers low. It is critical to perform the transplant within a few days of the last plasmapheresis because rebound of anti-HLA does occur and can negate the benefits achieved with previous treatments. The Mayo Clinic has adopted both the PP/low-dose IVIg and high-dose IVIg protocols with similar success (13,25). Table 2 shows the advantages and limitations of the high-dose IVIg versus PP/CMV-Ig protocols. Table 3 compares the results from the two protocols over the last 3 to 5 yr. As can be seen, outcomes for patients at the three institutions (JHH, Cedars-Sinai, and Mayo Clinic) are very similar and comparable to results from nonsensitized patients.

View this table:
  • View inline
  • View popup
Table 2.

High-dose IVIg versus PP + CMV-Ig: Advantages and limitationsa

View this table:
  • View inline
  • View popup
Table 3.

Long-term outcomes of desensitization protocols for highly sensitized patients with ESRDa

Complications and Cost of IVIg Therapy

Unlike the use in immunodeficiency, patients who are highly HLA sensitized require higher doses (1 to 2 g/kg per dose) to achieve a beneficial outcome. The use of higher doses and concentrations of IVIg products results in higher rates of infusion-related complications that, at first, were not anticipated and were poorly understood. We recently reviewed the complications associated with IVIg infusions in patients who had normal renal function and those who were on dialysis (23). Briefly, the safety of IVIg infusion (2 g/kg) doses given during a 4-h hemodialysis session monthly ×4 versus placebo (0.1% albumin) in equivalent doses was studied in the IGO2 trial (5). There were more than 300 infusions in each arm of the study using Gamimune N 10% versus placebo. Adverse events were similar in both arms of the study (24 IVIg versus 23 placebo). The most common adverse event in the IVIg arm was headache (52 versus 24%; P = 0.056). This usually abated with reduction in infusion rate and acetaminophen. Ten serious adverse events were noted; of these, nine were in the placebo group. Therefore, we concluded from this double-blind, placebo-controlled trial that high-dose IVIg infusions during hemodialysis are safe.

IVIg is an expensive therapy, and, ultimately, insurers and hospitals question the use of this drug for desensitization. Is it cost-effective? Data do exist in this regard (2,5). Currently, a four-dose course of IVIg for a 70-kg person at 2 g/kg would cost $30,000. However, one must compare this with the cost of maintaining patients on dialysis, which is the only other option. In the IGO2 study (5), the calculated cost savings was approximately $300,000 per patient who received a transplant versus those who remained on dialysis for the 5 yr of the study. Data from the U.S. Renal Data System (2003) also confirm that a considerable cost savings to Medicare is seen in highly sensitized patients who receive a transplant versus those who remain on dialysis (3).

Other Approaches to Desensitization

Mycophenolate mofetil (MMF) has been shown to decrease response to neoantigens in the transplant population (31). Min et al. (32) combined MMF with immunoabsorption and tacrolimus for the treatment of acute AMR. Six of 185 cadaver renal allograft recipients developed C4d-positive AMR approximately 5 d after transplant. After treatment, the PRA fell from values as high as 50 to 8% with resolution of the rejection in four of the six patients. Which of the components were responsible for this benefit could not be determined with certainty. In a study of pediatric heart recipients, Jacobs et al. (33) used this modality in the pretransplantation setting. In combination with IVIg and plasmapheresis, they were able to provide transplants to eight children who had an elevated PRA; however, the survival rate was only 50%. They concluded that these sensitized patients remain at high risk despite aggressive immunosuppression. In a study of 388 kidney transplant recipients who had a kidney biopsy within the first 6 mo after transplantation, C4d-positive rejection could be reduced by 50% when calcineurin inhibitor or MMF therapy was started 2 to 4 h before transplantation when compared with initiation after surgery (adjusted odds ratio 0.5; P = 0.03).

Glotz et al. (34) reported on the successful use of high-dose IVIg for desensitization. Removal of donor-specific antibody seems to be effective in allowing transplantation in highly sensitized patients and in treating AMR episodes. This first was reported by Pascual et al. (35), who combined plasma exchange with Cellcept and tacrolimus for treatment of AMR. Lehrich et al. (36) reported that IVIg combined with plasmapheresis is extremely effective in reversing AMR episodes. Schweitzer et al. (37) reported on a protocol using plasma exchange to improve transplant rates in highly sensitized patients. Zand et al. (38) reported in vitro data that suggest that Thymoglobulin has significant effects on B cells and plasma cells in vitro and potentially could be useful as a desensitization agent. These approaches are limited by low numbers and/or no clinical experience but could be developed further if rigorous clinical trials are designed.

Although many variations on the theme of IVIg and plasma exchange have been presented, it is clear that both therapies have a place in the treatment of highly sensitized patients. Many possible combinations of therapy might be considered but should be undertaken with great caution as the risks for this patient population are high and the possibilities for repeat transplantation are few.

Sirolimus has similarly been noted to have anti-B cell activity with reduction of antibody formation in transplant patients (39), an effect that seems to be synergistic to that of IVIg (40). Toyoda et al. (40) suggested that lower concentration in combination with IVIg could represent an effective immunomodulatory drug combination.

Campath-1H (alemtuzumab) is a humanized mAb against CD52, a small glycosylphosphatidylinositol-anchored glycoprotein determinant that is highly expressed on both T and B cells (41). However, despite its reactivity against B cells, it has been unable to prevent and in fact may result in increased rates of C4d-positive, AMR when used without a calcineurin inhibitor (42–44). It also seems that memory T cells are resistant to alemtuzumab therapy (45,46). These findings would question its addition to desensitization protocols.

Protein A column immunoabsorption has been used primarily as part of both ABO incompatible transplant (47) and in the setting of allosensitized patients (48). Lorenz et al. (48) reported their 10-yr experience with 40 cadaver transplant recipients who had median PRA of 77% (nine of whom had a flow-positive CMX) and were treated with one absorption before and several after transplantation. Three-year graft survival was 71%, with 11% immunologic loss. Although the current dogma is that protein A columns deplete circulating Ig, Goodyear and Silverman (49,50), using murine models, hypothesized that the benefit comes from protein A that leaches off the column into the patient. They found that infusions of small quantities of protein A into mice led to profound and prolonged depletion of circulating and tissue B cells. Silverman and colleagues (49,51) suggested that perhaps direct administration of protein A may be beneficial for certain antibody-mediated diseases, akin to what is currently being tested with anti-CD20 therapy.

Rituximab, a chimeric (Figure 5) anti-CD20 (Figure 6) mAb that is approved for treatment of lymphoma, efficiently eliminates B cells (52). CD20 is expressed early in B cell ontogeny, but expression is absent on plasma cells (Figure 7). Rituximab eliminates B cells by three potential mechanisms: Antibody dependent, cell-mediated cytotoxicity; complement-dependent cytotoxicity; and apoptosis (Figure 8). In a study of patients who had end-stage renal failure, we found that rituximab could be given safely and at doses lower than those recommended for cancer treatment (53). Dosing was associated with release of TNF-α and had minimal effect on the ex vivo T cell immune responses (54). The B cells that recover are substantially depleted of memory CD27+ B cells for as long as 2 yr after a single dose (55). Dosing dramatically impairs response to a neoantigen given at the time of rituximab dosing but ultimately has no effect on preexisting memory responses (56). Last, we reported methods to perform CMX and PRA determinations in patients who had been treated with rituximab either by elimination of the cell surface CD20 by pronase treatment of the cells or by immunomagnetic bead absorption of the serum-containing rituximab (57,58).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Rituximab is a chimeric mAb that is similar to basiliximab. The antigen-combining portion is composed of murine-derived variable regions that are attached to human constant regions.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

CD20 is a membrane-bound molecule with two small separate extracellular domains and three separate intracellular domains.

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

CD20 expression first is weakly detectable at the immature B cell stage. Expression fades again at the plasma cell stage. It is expressed to the greatest amount on germinal center (GC) B cells.

Figure 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 8.

Rituximab is postulated to eliminate B cells by one of three mechanisms: Antibody-dependent cell-mediated cytotoxicity (ADCC), complement-mediated cytotoxicity, or activation of the apoptotic pathways.

In our small phase 1 study, we found that there was a decrease in PRA and or antibody specificity in most of the patients, data that were confirmed recently using single-antigen HLA beads (59). Most other reports also are small trials and focus on rituximab in ABO-incompatible transplants of either kidney (60) or liver (61,62) and as a method of sparing splenectomy (63,64). The largest experience is of 11 patients in whom rituximab was combined with immunoabsorption and IVIg without splenectomy and resulted in normal renal function in all 11 (64). However, a more recent report questioned the need for rituximab in such transplants (65). This highlights the major problem with rituximab: The absence of controlled clinical trials. Future mechanistic studies will be facilitated by the recent generation of a mouse strain that expresses human CD20 in a normally regulated manner on B cells (66). These animals, when treated with rituximab, undergo B cell elimination in a manner analogous to what is seen with rituximab treatment in humans. Future clinical trials might be facilitated by a humanized anti-CD20, 2H7 (Figure 5), now in early clinical trials for treatment of rheumatoid arthritis.

Alternative, Nonpharmacologic Approaches to Improve Transplantation Rates in Highly HLA-Sensitized Patients

As more transplant centers in the United States and around the world develop protocols to improve transplantation for the highly HLA-sensitized patients, other approaches have emerged. Claas et al. (67) reported on The Acceptable Mismatch Program, which has been developed for allocating kidneys to highly sensitized patients. These investigators reported a schema that was developed for Eurotransplant using a computer program, HLA Matchmaker, that allocates kidneys to patients on the basis of avoidance of antigen sensitization. The authors reported that 112 transplants had been performed with a 2-yr graft survival of 87% but gave no data on the incidence and the severity of rejection episodes or current serum creatinine values. They also suggested that this could be implemented in conjunction with desensitization protocols in an effort to provide transplants to the most highly sensitized patients. Other potential protocols include donor exchange programs that may improve access of highly sensitized patients to transplantation (59). If these approaches are successful in the United States, then they should be tested before initiation of desensitization therapy because of the reduced expense.

Conclusion

On the basis of multiple observations of the in vitro and in vivo effectiveness of IVIg in modulation of anti-HLA antibodies, the experience in the use of IVIg in inflammatory and autoimmune disorders, and the use in the treatment of severe AMR episodes in cardiac and renal allograft recipients (5,9–12,15), we believe that IVIg alone or in combination with PP has an important role in the treatment of highly sensitized patients who are awaiting transplantation. However, newer agents and combinations of agents are being tried with early evidence of anecdotal success. Clearly, large multicenter, well-controlled trials are needed in this field to provide optimal benefit to these patients who are otherwise unlikely to receive a transplant unless therapeutic interventions are undertaken to reduce anti-HLA antibodies.

Footnotes

  • Published online ahead of print. Publication date available at www.cjasn.org.

  • Copyright © 2006 by the American Society of Nephrology

References

  1. ↵
    Organ Procurement Transplantation Network/Scientific Registry of Transplant Recipients: 2004 Annual Report: Transplant Data 1994–2003, Department of Health and Human Services, Health Resources and Services Administration, Healthcare Systems Bureau, Division of Transplantation; United Network for Organ Sharing; University Renal Research and Education Association,2004
  2. ↵
    Jordan S, Cunningham-Rundles C, McEwan R: Utility of intravenous immune globulin in kidney transplantation: Efficacy, safety, and cost implications. Am J Transplant3 :653– 664,2003
    OpenUrlCrossRefPubMed
  3. ↵
    US Renal Data System: USRDS 2003 Annual Data Report; Atlas of End-Stage Renal Diseases in the United States, Bethesda, National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases,2003
  4. ↵
    Patel R, Terasaki PI: Significance of the positive crossmatch test in kidney transplantation. N Engl J Med280 :735– 739,1969
    OpenUrlCrossRefPubMed
  5. ↵
    Jordan SC, Tyan D, Stablein D, McIntosh M, Rose S, Vo A, Toyoda M, Davis C, Shapiro R, Adey D, Milliner D, Graff R, Steiner R, Ciancio G, Sahney S, Light J: Evaluation of intravenous immunoglobulin as an agent to lower allosensitization and improve transplantation in highly sensitized adult patients with end-stage renal disease: Report of the NIH IGO2 trial. J Am Soc Nephrol15 :3256– 3262,2004
    OpenUrlAbstract/FREE Full Text
  6. Meier-Kriesche HU, Port FK, Ojo AO, Rudich SM, Hanson JA, Cibrik DM, Leichtman AB, Kaplan B: Effect of waiting time on renal transplant outcome. Kidney Int58 :1311– 1317,2000
    OpenUrlCrossRefPubMed
  7. ↵
    Kaplan B, Schold J, Meier-Kriesche HU: Overview of large database analysis in renal transplantation. Am J Transplant3 :1052– 1056,2003
    OpenUrlCrossRefPubMed
  8. ↵
    Montgomery RA, Zachary AA, Racusen LC, Leffell MS, King KE, Burdick J, Maley WR, Ratner LE: Plasmapheresis and intravenous immunoglobulin provides effective rescue therapy for refractory humoral rejection and allows kidneys to be successfully transplanted into cross-match-positive recipients. Transplantation70 :887– 895,2000
    OpenUrlCrossRefPubMed
  9. ↵
    Tyan DB, Li VA, Czer L, Trento A, Jordan SC: Intravenous immunoglobulin suppression of HLA alloantibody in highly sensitized transplant candidates and transplantation with a histoincompatible organ. Transplantation57 :553– 562,1994
    OpenUrlCrossRefPubMed
  10. ↵
    Jordan SC, Quartel AW, Czer LS, Admon D, Chen G, Fishbein MC, Schwieger J, Steiner RW, Davis C, Tyan DB: Post-transplant therapy using high-dose human immunoglobulin (intravenous gammaglobulin) to control acute humoral rejection in renal and cardiac allograft recipients and potential mechanism of action. Transplantation66 :800– 805,1998
    OpenUrlCrossRefPubMed
  11. Jordan SC, Vo A, Bunnapradist S, Toyoda M, Peng A, Puliyanda D, Kamil E, Tyan D: Intravenous immune globulin treatment inhibits crossmatch positivity and allows for successful transplantation of incompatible organs in living-donor and cadaver recipients. Transplantation76 :631– 636,2003
    OpenUrlCrossRefPubMed
  12. ↵
    Jordan S, Tyan D: Intravenous gamma globulin (IVIG) inhibits lymphocytotoxic antibody in vitro [Abstract]. J Am Soc Nephrol2 :803 ,1991
    OpenUrl
  13. ↵
    Dean PG, Gloor JM, Stegall MD: Conquering absolute contraindications to transplantation: Positive-crossmatch and ABO-incompatible kidney transplantation. Surgery137 :269– 273,2005
    OpenUrlCrossRefPubMed
  14. ↵
    Kazatchkine MD, Kaveri SV: Immunomodulation of autoimmune and inflammatory diseases with intravenous immune globulin. N Engl J Med345 :747– 755,2001
    OpenUrlCrossRefPubMed
  15. ↵
    Toyoda M, Pao A, Petrosian A, Jordan SC: Pooled human gammaglobulin modulates surface molecule expression and induces apoptosis in human B cells. Am J Transplant3 :156– 166,2003
    OpenUrlCrossRefPubMed
  16. ↵
    Bayry J, Lacroix-Desmazes S, Carbonneil C, Misra N, Donkova V, Pashov A, Chevailler A, Mouthon L, Weill B, Bruneval P, Kazatchkine MD, Kaveri SV: Inhibition of maturation and function of dendritic cells by intravenous immunoglobulin. Blood101 :758– 765,2003
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Magee JC, Collins BH, Harland RC, Lindman BJ, Bollinger RR, Frank MM, Platt JL: Immunoglobulin prevents complement-mediated hyperacute rejection in swine-to-primate xenotransplantation. J Clin Invest96 :2404– 2412,1995
    OpenUrlCrossRefPubMed
  18. ↵
    Lutz HU, Stammler P, Bianchi V, Trueb RM, Hunziker T, Burger R, Jelezarova E, Spath PJ: Intravenously applied IgG stimulates complement attenuation in a complement-dependent autoimmune disease at the amplifying C3 convertase level. Blood103 :465– 472,2004
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Sameulsson A, Towers TL, Ravetch JV: Anti-inflamatory activity of IvIG mediated through the inhibitory Fc receptor. Science29 :484– 486,2001
    OpenUrlCrossRef
  20. ↵
    Bohmig GA, Exner M, Habicht A, Schillinger M, Lang U, Kletzmayr J, Saemann MD, Horl WH, Watschinger B, Regele H: Capillary C4d deposition in kidney allografts: A specific marker of alloantibody-dependent graft injury. J Am Soc Nephrol13 :1091– 1099,2002
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Williams JM, Holzknecht ZE, Plummer TB, Lin SS, Brunn GJ, Platt JL: Acute vascular rejection and accommodation: Divergent outcomes of the humoral response to organ transplantation. Transplantation78 :1471– 1478,2004
    OpenUrlCrossRefPubMed
  22. ↵
    Abe J, Jibiki T, Noma S, Nakajima T, Saito H, Terai M: Gene expression profiling of the effect of high-dose intravenous Ig in patients with Kawasaki disease. J Immunol174 :5837– 5845,2005
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Gill V, Doig C, Knight D, Love E, Kubes P: Targeting adhesion molecules as a potential mechanism of action for intravenous immunoglobulin. Circulation112 :2031– 2039,2005
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Wassmuth R, Hauser IA, Schuler K, Erxleben H, Arnold ML, Koelman CA, Claas FH, Kalden JR: Differential inhibitory effects of intravenous immunoglobulin preparations on HLA-alloantibodies in vitro. Transplantation71 :1436– 1442,2001
    OpenUrlCrossRefPubMed
  25. ↵
    Gloor JM, DeGoey S, Ploeger N, Gebel H, Bray R, Moore SB, Dean PG, Stegall MD: Persistence of low levels of alloantibody after desensitization in crossmatch-positive living-donor kidney transplantation. Transplantation78 :221– 227,2004
    OpenUrlPubMed
  26. Luke PP, Scantlebury VP, Jordan ML, Vivas CA, Hakala TR, Jain A, Somani A, Fedorek S, Randhawa P, Shapiro R: Reversal of steroid- and anti-lymphocyte antibody-resistant rejection using intravenous immunoglobulin (IVIG) in renal transplant recipients. Transplantation72 :419– 422,2001
    OpenUrlCrossRefPubMed
  27. Glotz D, Antoine C, Julia P, Pegaz-Fiornet B, Duboust A, Boudjeltia S, Fraoui R, Combes M, Bariety J: Intravenous immunoglobulins and transplantation for patients with anti-HLA antibodies. Transpl Int17 :1– 8,2004
    OpenUrlCrossRefPubMed
  28. ↵
    De Marco T, Damon LE, Colombe B, Keith F, Chatterjee K, Garovoy MR: Successful immunomodulation with intravenous gamma globulin and cyclophosphamide in an alloimmunized heart transplant recipient. J Heart Lung Transplant16 :360– 365,1997
    OpenUrlPubMed
  29. ↵
    Casadei DH, del C Rial M, Opelz G, Golberg JC, Argento JA, Greco G, Guardia OE, Haas E, Raimondi EH: A randomized and prospective study comparing treatment with high-dose intravenous immunoglobulin with monoclonal antibodies for rescue of kidney grafts with steroid-resistant rejection. Transplantation71 :53– 58,2001
    OpenUrlCrossRefPubMed
  30. ↵
    Montgomery RA, Zachary AA: Transplanting patients with a positive donor-specific crossmatch: A single center’s perspective. Pediatr Transplant8 :535– 542,2004
    OpenUrlCrossRefPubMed
  31. ↵
    Kimball J, Pescovitz MD, Book BK, Norman D: Mycophenolate mofetil reduces human IgG anti-ATGAM antibody formation. Transplantation60 :1379– 1383,1995
    OpenUrlPubMed
  32. ↵
    Min L, Shuming J, Zheng T, Daxi J, Huiping C, Zhihong L, Leishi L: Novel rescue therapy for C4d-positive acute humoral renal allograft rejection. Clin Transplant19 :51– 55,2005
    OpenUrlCrossRefPubMed
  33. ↵
    Jacobs JP, Quintessenza JA, Boucek RJ, Morell VO, Botero LM, Badhwar V, van Gelder HM, Asante-Korang A, McCormack J, Daicoff GR: Pediatric cardiac transplantation in children with high panel reactive antibody. Ann Thorac Surg78 :1703– 1709,2004
    OpenUrlCrossRefPubMed
  34. ↵
    Glotz D, Antoine C, Julia P, Suberbielle-Boissel C, Boudjeltia S, Fraoui R, Hacen C, Duboust A, Bariety J: Desensitization and subsequent kidney transplantation of patients using intravenous immunoglobulins (IVIg). Am J Transplant2 :758– 760,2002
    OpenUrlCrossRefPubMed
  35. ↵
    Pascual M, Saidman S, Tolkoff-Rubin N, Williams WW, Mauiyyedi S, Duan JM, Farrell ML, Colvin RB, Cosimi AB, Delmonico FL: Plasma exchange and tacrolimus-mycophenolate rescue for acute humoral rejection in kidney transplantation. Transplantation66 :1460– 1464,1998
    OpenUrlCrossRefPubMed
  36. ↵
    Lehrich RW, Rocha PN, Reinsmoen N, Greenberg A, Butterly DW, Howell DN, Smith SR: Intravenous immunoglobulin and plasmapheresis in acute humoral rejection: Experience in renal allograft transplantation. Hum Immunol66 :350– 358,2005
    OpenUrlCrossRefPubMed
  37. ↵
    Schweitzer EJ, Wilson JS, Fernandez-Vina M, Fox M, Gutierrez M, Wiland A, Hunter J, Farney A, Philosophe B, Colonna J, Jarrell BE, Bartlett ST: A high panel-reactive antibody rescue protocol for cross-match-positive live donor kidney transplants. Transplantation70 :1531– 1536,2000
    OpenUrlCrossRefPubMed
  38. ↵
    Zand MS, Vo T, Huggins J, Felgar R, Liesveld J, Pellegrin T, Bozorgzadeh A, Sanz I, Briggs BJ: Polyclonal rabbit antithymocyte globulin triggers B-cell and plasma cell apoptosis by multiple pathways. Transplantation79 :1507– 1515,2005
    OpenUrlCrossRefPubMed
  39. ↵
    Pescovitz MD, Book BK, Henson S, Leapman SB, Milgrom ML, Kimball J, Norman D, Filo RS: The addition of sirolimus to cyclosporine and steroids inhibits the anti-equine antibody response in renal transplant recipients treated with antithymocyte globulin. Am J Transplant3 :497– 500,2003
    OpenUrlCrossRefPubMed
  40. ↵
    Toyoda M, Petrosyan A, Pao A, Jordan SC: Immunomodulatory effects of combination of pooled human gammaglobulin and rapamycin on cell proliferation and apoptosis in the mixed lymphocyte reaction. Transplantation78 :1134– 1138,2004
    OpenUrlCrossRefPubMed
  41. ↵
    Ravandi F, O’Brien S: Alemtuzumab. Expert Rev Anticancer Ther5 :39– 51,2005
    OpenUrlCrossRefPubMed
  42. ↵
    Hill P, Gagliardini E, Ruggenenti P, Remuzzi G: Severe early acute humoral rejection resulting in allograft loss in a renal transplant recipient with Campath-1H induction therapy. Nephrol Dial Transplant20 :1741– 1744,2005
    OpenUrlCrossRefPubMed
  43. Cai J, Terasaki PI, Bloom DD, Torrealba JR, Friedl A, Sollinger HW, Knechtle SJ: Correlation between human leukocyte antigen antibody production and serum creatinine in patients receiving sirolimus monotherapy after Campath-1H induction. Transplantation78 :919– 924,2004
    OpenUrlCrossRefPubMed
  44. ↵
    Bartosh SM, Knechtle SJ, Sollinger HW: Campath-1H use in pediatric renal transplantation. Am J Transplant5 :1569– 1573,2005
    OpenUrlCrossRefPubMed
  45. ↵
    Pearl JP, Parris J, Hale DA, Hoffmann SC, Bernstein WB, McCoy KL, Swanson SJ, Mannon RB, Roederer M, Kirk AD: Immunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletion. Am J Transplant5 :465– 474,2005
    OpenUrlCrossRefPubMed
  46. ↵
    Kirk AD, Hale DA, Mannon RB, Kleiner DE, Hoffmann SC, Kampen RL, Cendales LK, Tadaki DK, Harlan DM, Swanson SJ: Results from a human renal allograft tolerance trial evaluating the humanized CD52-specific monoclonal antibody alemtuzumab (CAMPATH-1H). Transplantation76 :120– 129,2003
    OpenUrlCrossRefPubMed
  47. ↵
    Squifflet JP, De Meyer M, Malaise J, Latinne D, Pirson Y, Alexandre GP: Lessons learned from ABO-incompatible living donor kidney transplantation: 20 years later. Exp Clin Transplant2 :208– 213,2004
    OpenUrlPubMed
  48. ↵
    Lorenz M, Regele H, Schillinger M, Kletzmayr J, Haidbauer B, Derfler K, Druml W, Bohmig GA: Peritransplant immunoadsorption: A strategy enabling transplantation in highly sensitized crossmatch-positive cadaveric kidney allograft recipients. Transplantation79 :696– 701,2005
    OpenUrlCrossRefPubMed
  49. ↵
    Goodyear CS, Silverman GJ: B cell superantigens: A microbe’s answer to innate-like B cells and natural antibodies. Springer Semin Immunopathol26 :463– 484,2005
    OpenUrlCrossRefPubMed
  50. ↵
    Goodyear CS, Silverman GJ: Staphylococcal toxin induced preferential and prolonged in vivo deletion of innate-like B lymphocytes. Proc Natl Acad Sci U S A101 :11392– 11397,2004
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Silverman GJ, Goodyear CS, Siegel DL: On the mechanism of staphylococcal protein A immunomodulation. Transfusion45 :274– 280,2005
    OpenUrlCrossRefPubMed
  52. ↵
    Pescovitz MD: The use of rituximab, anti-CD20 monoclonal antibody, in pediatric transplantation. Pediatr Transplant8 :9– 21,2004
    OpenUrlCrossRefPubMed
  53. ↵
    Vieira CA, Agarwal A, Book BK, Sidner RA, Bearden CM, Gebel HM, Roggero AL, Fineberg NS, Taber T, Kraus MA, Pescovitz MD: Rituximab for reduction of anti-HLA antibodies in patients awaiting renal transplantation: 1. Safety, pharmacodynamics, and pharmacokinetics. Transplantation77 :542– 548,2004
    OpenUrlCrossRefPubMed
  54. ↵
    Agarwal A, Vieira CA, Book BK, Sidner RA, Fineberg NS, Pescovitz MD: Rituximab, anti-CD20, induces in vivo cytokine release but does not impair ex vivo T-cell responses. Am J Transplant4 :1357– 1360,2004
    OpenUrlCrossRefPubMed
  55. ↵
    Sidner RA, Book BK, Agarwal A, Bearden CM, Vieira CA, Pescovitz MD: In vivo human B-cell subset recovery after in vivo depletion with rituximab, anti-human CD20 monoclonal antibody. Hum Antibodies13 :55– 62,2004
    OpenUrlPubMed
  56. ↵
    Bearden CM, Agarwal A, Book BK, Vieira CA, Sidner RA, Ochs HD, Young M, Pescovitz MD: Rituximab inhibits the in vivo primary and secondary antibody response to a neoantigen, bacteriophage phiX174. Am J Transplant5 :50– 57,2005
    OpenUrlCrossRefPubMed
  57. ↵
    Book BK, Agarwal A, Milgrom AB, Bearden CM, Sidner RA, Higgins NG, Pescovitz MD: New crossmatch technique eliminates interference by humanized and chimeric monoclonal antibodies. Transplant Proc37 :640– 642,2005
    OpenUrlCrossRefPubMed
  58. ↵
    Bearden CM, Book BK, Sidner RA, Pescovitz MD: Removal of therapeutic anti-lymphocyte antibodies from human sera prior to anti-human leukocyte antibody testing. J Immunol Methods300 :192– 199,2005
    OpenUrlCrossRefPubMed
  59. ↵
    Kaplan I, Houp JA, Leffell MS, Hart JM, Zachary AA: A computer match program for paired and unconventional kidney exchanges. Am J Transplant5 :2306– 2308,2005
    OpenUrlCrossRefPubMed
  60. ↵
    Sawada T, Fuchinoue S, Kawase T, Kubota K, Teraoka S: Preconditioning regimen consisting of anti-CD20 monoclonal antibody infusions, splenectomy and DFPP-enabled non-responders to undergo ABO-incompatible kidney transplantation. Clin Transplant18 :254– 260,2004
    OpenUrlCrossRefPubMed
  61. ↵
    Kawagishi N, Satoh K, Enomoto Y, Akamatsu Y, Sekiguchi S, Fukumori T, Fujimori K, Satomi S: New strategy for ABO-incompatible living donor liver transplantation with anti-CD20 antibody (rituximab) and plasma exchange. Transplant Proc37 :1205– 1206,2005
    OpenUrlCrossRefPubMed
  62. ↵
    Usuda M, Fujimori K, Koyamada N, Fukumori T, Sekiguchi S, Kawagishi N, Akamatsu Y, Enomoto Y, Satoh K, Satoh A, Ishida K, Moriya T, Satomi S: Successful use of anti-CD20 monoclonal antibody (rituximab) for ABO-incompatible living-related liver transplantation. Transplantation79 :12– 16,2005
    OpenUrlCrossRefPubMed
  63. ↵
    Sonnenday CJ, Warren DS, Cooper M, Samaniego M, Haas M, King KE, Shirey RS, Simpkins CE, Montgomery RA: Plasmapheresis, CMV hyperimmune globulin, and anti-CD20 allow ABO-incompatible renal transplantation without splenectomy. Am J Transplant4 :1315– 1322,2004
    OpenUrlCrossRefPubMed
  64. ↵
    Tyden G, Kumlien G, Genberg H, Sandberg J, Lundgren T, Fehrman I: ABO incompatible kidney transplantations without splenectomy, using antigen-specific immunoadsorption and rituximab. Am J Transplant5 :145– 148,2005
    OpenUrlCrossRefPubMed
  65. ↵
    Gong Q, Ou Q, Ye S, Lee WP, Cornelius J, Diehl L, Lin WY, Hu Z, Lu Y, Chen Y, Wu Y, Meng YG, Gribling P, Lin Z, Nguyen K, Tran T, Zhang Y, Rosen H, Martin F, Chan AC: Importance of cellular microenvironment and circulatory dynamics in B cell immunotherapy. J Immunol174 :817– 826,2005
    OpenUrlAbstract/FREE Full Text
  66. ↵
    Segev DL, Simpkins CE, Warren DS, King KE, Shirey RS, Maley WR, Melancon JK, Cooper M, Kozlowski T, Montgomery RA: ABO incompatible high-titer renal transplantation without splenectomy or anti-CD20 treatment. Am J Transplant5 :2570– 2575,2005
    OpenUrlCrossRefPubMed
  67. ↵
    Claas FH, Witvliet MD, Duquesnoy RJ, Persijn GG, Doxiadis II: The acceptable mismatch program as a fast tool for highly sensitized patients awaiting a cadaveric kidney transplantation: Short waiting time and excellent graft outcome. Transplantation78 :190– 193,2004
    OpenUrlPubMed
PreviousNext
Back to top

In this issue

Clinical Journal of the American Society of Nephrology
Vol. 1, Issue 3
May 2006
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
View Selected Citations (0)
Print
Download PDF
Sign up for Alerts
Email Article
Thank you for your help in sharing the high-quality science in CJASN.
Enter multiple addresses on separate lines or separate them with commas.
Presensitization: The Problem and Its Management
(Your Name) has sent you a message from American Society of Nephrology
(Your Name) thought you would like to see the American Society of Nephrology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Presensitization: The Problem and Its Management
Stanley C. Jordan, Mark D. Pescovitz
CJASN May 2006, 1 (3) 421-432; DOI: 10.2215/CJN.01651105

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Presensitization: The Problem and Its Management
Stanley C. Jordan, Mark D. Pescovitz
CJASN May 2006, 1 (3) 421-432; DOI: 10.2215/CJN.01651105
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Mechanism of Action of IVIg
    • Clinical Use of IVIg in Kidney Transplantation
    • National Institutes of Health IGO2 Study
    • Plasmapheresis/CMV-Ig Protocol
    • Complications and Cost of IVIg Therapy
    • Other Approaches to Desensitization
    • Alternative, Nonpharmacologic Approaches to Improve Transplantation Rates in Highly HLA-Sensitized Patients
    • Conclusion
    • Footnotes
    • References
  • Figures & Data Supps
  • Info & Metrics
  • View PDF

More in this TOC Section

  • Recommended Clinical Trial End Points for Dialysis Catheters
  • Clinical Trial End Points for Hemodialysis Vascular Access
  • Definitions and End Points for Interventional Studies for Arteriovenous Dialysis Access
Show more Moving Points in Nephrology

Cited By...

  • Pretransplant Desensitization with Costimulation Blockade and Proteasome Inhibitor Reduces DSA and Delays Antibody-Mediated Rejection in Highly Sensitized Nonhuman Primate Kidney Transplant Recipients
  • Daratumumab in Sensitized Kidney Transplantation: Potentials and Limitations of Experimental and Clinical Use
  • Successful desensitization with proteasome inhibition and costimulation blockade in sensitized nonhuman primates
  • Late Kidney Dysfunction in a Kidney Transplant Recipient
  • High-Dosage Intravenous Immunoglobulin-Associated Macrovacuoles Are Associated with Chronic Tubulointerstitial Lesion Worsening in Renal Transplant Recipients
  • Google Scholar

Similar Articles

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Articles

  • Current Issue
  • Early Access
  • Subject Collections
  • Article Archive
  • ASN Meeting Abstracts

Information for Authors

  • Submit a Manuscript
  • Trainee of the Year
  • Author Resources
  • ASN Journal Policies
  • Reuse/Reprint Policy

About

  • CJASN
  • ASN
  • ASN Journals
  • ASN Kidney News

Journal Information

  • About CJASN
  • CJASN Email Alerts
  • CJASN Key Impact Information
  • CJASN Podcasts
  • CJASN RSS Feeds
  • Editorial Board

More Information

  • Advertise
  • ASN Podcasts
  • ASN Publications
  • Become an ASN Member
  • Feedback
  • Follow on Twitter
  • Password/Email Address Changes
  • Subscribe to ASN Journals

© 2022 American Society of Nephrology

Print ISSN - 1555-9041 Online ISSN - 1555-905X

Powered by HighWire