Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Blood Adv ; 8(12): 3001-3012, 2024 Jun 25.
Article En | MEDLINE | ID: mdl-38625984

ABSTRACT: Patients with Epstein-Barr virus (EBV)-positive posttransplant lymphoproliferative disease (EBV+ PTLD) in whom initial treatment fails have few options and historically low median overall survival (OS) of 0.7 months after allogeneic hematopoietic cell transplant (HCT) and 4.1 months after solid organ transplant (SOT). Tabelecleucel is an off-the-shelf, allogeneic EBV-specific cytotoxic T-lymphocyte immunotherapy for EBV+ PTLD. Previous single-center experience showed responses in patients with EBV+ PTLD after HCT or SOT. We now report outcomes from a multicenter expanded access protocol in HCT (n = 14) and SOT (n = 12) recipients treated with tabelecleucel for EBV+ PTLD that was relapsed/refractory (R/R) to rituximab with/without chemotherapy. The investigator-assessed objective response rate was 65.4% overall (including 38.5% with a complete and 26.9% with a partial response), 50.0% in HCT, and 83.3% in SOT. The estimated 1- and 2-year OS rates were both 70.0% (95% confidence interval [CI], 46.5-84.7) overall, both 61.5% (95% CI, 30.8-81.8) in HCT, and both 81.5% (95% CI, 43.5-95.1) in SOT (median follow-up: 8.2, 2.8, and 22.5 months, respectively). Patients responding to tabelecleucel had higher 1- and 2-year OS rates (94.1%) than nonresponders (0%). Treatment was well tolerated, with no reports of tumor flare, cytokine release syndrome, or rejection of marrow and SOT. Results demonstrate clinically meaningful outcomes across a broad population treated with tabelecleucel, indicating a potentially transformative and accessible treatment advance for R/R EBV+ PTLD after HCT or SOT. This trial was registered at www.ClinicalTrials.gov as #NCT02822495.


Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Lymphoproliferative Disorders , Transplantation, Homologous , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/therapy , Female , Male , Adult , Middle Aged , Herpesvirus 4, Human , Organ Transplantation/adverse effects , Aged , Treatment Outcome , Young Adult
2.
Lancet Oncol ; 25(3): 376-387, 2024 Mar.
Article En | MEDLINE | ID: mdl-38309282

BACKGROUND: Survival in Epstein-Barr virus (EBV)-positive post-transplant lymphoproliferative disease following haematopoietic stem-cell transplant (HSCT) or solid organ transplant (SOT) is poor after failure of initial therapy, indicating an urgent need for therapies for this ultra-rare disease. With recent EU marketing authorisation, tabelecleucel is the first off-the-shelf, allogeneic, EBV-specific T-cell immunotherapy to receive approval for treatment of relapsed or refractory EBV-positive post-transplant lymphoproliferative disease. We aimed to determine the clinical benefit of tabelecleucel in patients with relapsed or refractory EBV-positive post-transplant lymphoproliferative disease following HSCT or SOT. METHODS: In this global, multicentre, open-label, phase 3 trial, eligible patients (of any age) had biopsy-proven EBV-positive post-transplant lymphoproliferative disease, disease that was relapsed or refractory to rituximab after HSCT and rituximab with or without chemotherapy after SOT, and partially HLA-matched and appropriately HLA-restricted tabelecleucel available. Patients received tabelecleucel administered intravenously at 2 × 106 cells per kg on days 1, 8, and 15 in 35-day cycles and are assessed for up to 5 years for survival post-treatment initiation. The primary endpoint was objective response rate. All patients who received at least one dose of tabelecleucel were included in safety and efficacy analyses. This trial is registered with ClinicalTrials.gov, NCT03394365, and is ongoing. FINDINGS: From June 27, 2018, to Nov 5, 2021, 63 patients were enrolled, of whom 43 (24 [56%] male and 19 [44%] female) were included, 14 had prior HSCT, 29 had SOT. Seven (50%, 95% CI 23-77) of 14 participants in the HSCT group and 15 (52%, 33-71) of 29 participants in the SOT group had an objective response, with a median follow-up of 14·1 months (IQR 5·7-23·9) and 6·0 months (1·8-18·4), respectively. The most common grade 3 or 4 treatment-emergent adverse events were disease progression (in four [29%] of 14 in HSCT and eight [28%] of 29 in SOT) and decreased neutrophil count (in four [29%] of 14 in HSCT and four [14%] of 29 in SOT). Treatment-emergent serious adverse events were reported in 23 (53%) of 43 patients and fatal treatment-emergent adverse events in five (12%); no fatal treatment-emergent adverse event was treatment-related. There were no reports of tumour flare reaction, cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, transmission of infectious diseases, marrow rejection, or infusion reactions. No events of graft-versus-host disease or SOT rejection were reported as related to tabelecleucel. INTERPRETATION: Tabelecleucel provides clinical benefit in patients with relapsed or refractory EBV-positive post-transplant lymphoproliferative disease, for whom there are no other approved therapies, without evidence of safety concerns seen with other adoptive T-cell therapies. These data represent a potentially transformative and accessible treatment advance for patients with relapsed or refractory disease with few treatment options. FUNDING: Atara Biotherapeutics.


Epstein-Barr Virus Infections , Hematopoietic Stem Cell Transplantation , Lymphoproliferative Disorders , Organ Transplantation , Humans , Male , Female , Rituximab/adverse effects , Herpesvirus 4, Human/genetics , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/etiology , Alleles , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/etiology , Organ Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects
3.
Endocrinol Metab (Seoul) ; 36(1): 60-69, 2021 02.
Article En | MEDLINE | ID: mdl-33677928

BACKGROUND: This phase 3 study evaluated the efficacy and safety of 6-month treatment with romosozumab in Korean postmenopausal women with osteoporosis. METHODS: Sixty-seven postmenopausal women with osteoporosis (bone mineral density [BMD] T-scores ≤-2.5 at the lumbar spine, total hip, or femoral neck) were randomized (1:1) to receive monthly subcutaneous injections of romosozumab (210 mg; n=34) or placebo (n=33) for 6 months. RESULTS: At month 6, the difference in the least square (LS) mean percent change from baseline in lumbar spine BMD (primary efficacy endpoint) between the romosozumab (9.5%) and placebo (-0.1%) groups was significant (9.6%; 95% confidence interval, 7.6 to 11.5; P<0.001). The difference in the LS mean percent change from baseline was also significant for total hip and femoral neck BMD (secondary efficacy endpoints). After treatment with romosozumab, the percent change from baseline in procollagen type 1 N-terminal propeptide transiently increased at months 1 and 3, while that in C-terminal telopeptide of type 1 collagen showed a sustained decrease. No events of cancer, hypocalcemia, injection site reaction, positively adjudicated atypical femoral fracture or osteonecrosis of the jaw, or positively adjudicated serious cardiovascular adverse events were observed. At month 9, 17.6% and 2.9% of patients in the romosozumab group developed binding and neutralizing antibodies, respectively. CONCLUSION: Treatment with romosozumab for 6 months was well tolerated and significantly increased lumbar spine, total hip, and femoral neck BMD compared with placebo in Korean postmenopausal women with osteoporosis (ClinicalTrials.gov identifier NCT02791516).


Bone Density Conservation Agents , Osteoporosis , Antibodies, Monoclonal , Bone Density , Bone Density Conservation Agents/adverse effects , Female , Humans , Osteoporosis/chemically induced , Postmenopause , Republic of Korea
4.
J Bone Miner Metab ; 38(4): 605, 2020 07.
Article En | MEDLINE | ID: mdl-32198616

In the original publication of the article, the last row of Table 1 was published incorrectly as "Serum P1NP (µmol/L), median (IQR)b : Romosozumab, 25 (18, 34); Teriparatide, 25 (20, 33)". The correct row should be read as "Serum P1NP (µg/L), median (IQR)b : Romosozumab, 25 (18, 34); Teriparatide, 25 (20, 33)".

5.
J Bone Miner Metab ; 38(3): 310-315, 2020 May.
Article En | MEDLINE | ID: mdl-31707465

INTRODUCTION: Procollagen type I N-terminal propeptide (P1NP), a bone formation marker, reportedly predicts bone mineral density (BMD) response to teriparatide treatment in treatment-naive patients with osteoporosis. Results from a randomized, phase 3, open-label, active-controlled trial- STRUCTURE-showed that in patients previously treated with bisphosphonates, romosozumab led to gains in hip BMD, which were not observed with teriparatide. This post hoc analysis investigated the comparative utility of early changes in P1NP in predicting BMD response in patients who participated in the STRUCTURE trial, which enrolled patients who switched treatment from bisphosphonates to romosozumab/teriparatide. MATERIALS AND METHODS: Postmenopausal women (aged 55-90 years) with osteoporosis who had previously taken bisphosphonates were randomized to receive open-label subcutaneous romosozumab (210 mg once monthly; n = 218) or teriparatide (20 µg once daily; n = 218) for 12 months. BMD was assessed by dual-energy X-ray absorptiometry at the proximal femur and lumbar spine (LS) at baseline and months 6 and 12. To assess the utility of P1NP, the positive predictive value of increase from baseline in P1NP of > 10 µg/L at month 1 and achievement of various thresholds of percent change from baseline in BMD at month 12 were evaluated. RESULTS: Overall, 95% (191/202) of patients in the romosozumab group and 91% (183/201) in the teriparatide group demonstrated an increase in P1NP of > 10 µg/L from baseline at month 1. Among these patients, 18% and 3% of romosozumab-treated patients versus 60% and 12% of teriparatide-treated patients showed no increase from baseline (i.e., ≤ 0%) in total hip and LS BMD, respectively, at month 12. These data indicate that in patients switching from bisphosphonates to a bone-forming therapy, increases in P1NP do not help predict the hip BMD response. Although most patients treated with either teriparatide or romosozumab showed an increase in P1NP, the majority of patients on romosozumab showed an increase in hip BMD, while more than half of the patients on teriparatide did not. Teriparatide therapy did not increase total hip BMD in the majority of patients who transitioned from bisphosphonates to teriparatide. CONCLUSIONS: Thus, increases in P1NP were not predictive of BMD response in the teriparatide group because in approximately 60% of the patients who were administered teriparatide, the hip BMD decreased independent of the change in P1NP levels.


Alendronate/pharmacology , Antibodies, Monoclonal/pharmacology , Biomarkers/metabolism , Bone Density/physiology , Bone Remodeling/physiology , Peptide Fragments/metabolism , Procollagen/metabolism , Teriparatide/pharmacology , Aged , Aged, 80 and over , Bone Density/drug effects , Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Female , Humans , Middle Aged , Teriparatide/administration & dosage
6.
Arch Osteoporos ; 14(1): 59, 2019 06 05.
Article En | MEDLINE | ID: mdl-31168657

Romosozumab, which binds sclerostin, rebuilds the skeletal foundation before transitioning to antiresorptive treatment. This subgroup analysis of an international, randomized, double-blind study in postmenopausal women with osteoporosis showed efficacy and safety outcomes for romosozumab followed by denosumab in Japanese women were generally consistent with those for the overall population. PURPOSE: In the international, randomized, double-blind, phase 3 FRActure study, in postmenopausal woMen with ostEoporosis (FRAME; NCT01575834), romosozumab followed by denosumab significantly improved bone mineral density (BMD) and reduced fracture risk. This report evaluates Japanese women in FRAME. METHODS: Postmenopausal women with osteoporosis (T-score - 3.5 to - 2.5 at total hip or femoral neck) received romosozumab 210 mg or placebo subcutaneously monthly for 12 months, then each group received denosumab 60 mg subcutaneously every 6 months for 24 months. The key endpoint for Japanese women was BMD change. Other endpoints included new vertebral, clinical, and nonvertebral fracture; the subgroup analysis did not have adequate power to demonstrate statistically significant reductions. RESULTS: Of 7180 enrolled subjects, 492 (6.9%) were Japanese (247 romosozumab, 245 placebo). BMD increases from baseline were greater (P < 0.001) for romosozumab-to-denosumab than placebo-to-denosumab at the lumbar spine (36 months, 12.7%), total hip (4.2%), and femoral neck (4.1%). Fracture risk was lower through 36 months for romosozumab-to-denosumab vs placebo-to-denosumab for new vertebral (1.7% vs 4.5%; relative risk reduction (RRR) 63%, P = 0.070), clinical (3.2% vs 7.3%; RRR 53%, P = 0.072), nonvertebral (2.8% vs 6.1%; RRR 50%, P = 0.12), and all other fracture types evaluated. Rates of adverse events and positively adjudicated serious cardiovascular events were generally balanced between groups. CONCLUSIONS: Efficacy and safety for romosozumab-to-denosumab were similar between Japanese women and the overall population. The sequence of romosozumab to rebuild the skeletal foundation before transitioning to antiresorptive treatment with denosumab is a promising regimen for Japanese postmenopausal women with osteoporosis at high risk of fracture.


Antibodies, Monoclonal/administration & dosage , Bone Density Conservation Agents/administration & dosage , Bone Density/drug effects , Denosumab/administration & dosage , Osteoporosis, Postmenopausal/drug therapy , Aged , Aged, 80 and over , Double-Blind Method , Female , Femur Neck , Fractures, Bone/epidemiology , Fractures, Bone/prevention & control , Humans , Japan/epidemiology , Lumbar Vertebrae , Middle Aged , Osteoporosis , Risk , Risk Reduction Behavior
7.
J Bone Miner Res ; 34(3): 419-428, 2019 03.
Article En | MEDLINE | ID: mdl-30508316

Romosozumab, a humanized monoclonal antibody that binds and inhibits sclerostin, has the dual effect of increasing bone formation and decreasing bone resorption. As previously reported in the pivotal FRActure study in postmenopausal woMen with ostEoporosis (FRAME), women with a T-score of ≤ -2.5 at the total hip or femoral neck received subcutaneous placebo or romosozumab once monthly for 12 months, followed by open-label subcutaneous denosumab every 6 months for an additional 12 months. Upon completion of the 24-month primary analysis period, eligible women entered the extension phase and received denosumab for an additional 12 months. Here, we report the final analysis results through 36 months, including efficacy assessments of new vertebral, clinical, and nonvertebral fracture; bone mineral density (BMD); and safety assessments. Of 7180 women enrolled, 5743 (80%) completed the 36-month study (2851 romosozumab-to-denosumab; 2892 placebo-to-denosumab). Through 36 months, fracture risk was reduced in subjects receiving romosozumab versus placebo for 12 months followed by 24 months of denosumab for both groups: new vertebral fracture (relative risk reduction [RRR], 66%; incidence, 1.0% versus 2.8%; p < 0.001), clinical fracture (RRR, 27%; incidence, 4.0% versus 5.5%; p = 0.004), and nonvertebral fracture (RRR, 21%; incidence, 3.9% versus 4.9%; p = 0.039). BMD continued to increase for the 2 years with denosumab treatment in both arms. The substantial difference in BMD achieved through 12 months of romosozumab treatment versus placebo was maintained through the follow-up period when both treatment arms received denosumab. Subject incidence of adverse events, including positively adjudicated serious cardiovascular adverse events, were overall balanced between groups. In conclusion, in postmenopausal women with osteoporosis, 12 months of romosozumab led to persistent fracture reduction benefit and ongoing BMD gains when followed by 24 months of denosumab. The sequence of romosozumab followed by denosumab may be a promising regimen for the treatment of osteoporosis. © 2018 American Society for Bone and Mineral Research.


Antibodies, Monoclonal/therapeutic use , Denosumab/therapeutic use , Fracture Fixation , Fractures, Bone/drug therapy , Risk Reduction Behavior , Aged , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Biomarkers/metabolism , Bone Density/drug effects , Bone Remodeling/drug effects , Denosumab/adverse effects , Denosumab/pharmacology , Drug Administration Schedule , Drug Therapy, Combination , Fractures, Bone/epidemiology , Humans , Incidence
8.
Front Biosci (Elite Ed) ; 8(1): 1-14, 2016 01 01.
Article En | MEDLINE | ID: mdl-26709641

While has been considerable progress in the short-term outcomes following renal transplantation over the last several decades, minimal gains have been made with regards to long-term graft function and patient survival (1). The lack of long-term gains has been attributed to factors such as antibody mediated rejection (AMR), chronic allograft nephropathy (CAN), and toxicity to the allograft secondary to immunosuppression. Ischemia reperfusion injury (IRI) is also thought to contribute to poor long-term graft function, and its impact on patient and graft outcomes will likely expand with the increasing use of marginal kidneys secondary to organ shortages. While patient survival remains far below that of the general population, the causes of death have evolved in recent years with decreases in the rate of death from cardiovascular disease and infection, and increases secondary to malignancy (2), which are largely attributable to the potency of modern immunosuppression. As such, the development of novel therapies which can prevent delayed graft function (DGF), minimize AMR, while simultaneously reducing toxicity is vital to the improvement of long-term graft and patient outcomes.


Health Services Needs and Demand , Kidney Transplantation , Graft Rejection/immunology , Humans , Kidney Glomerulus/pathology , Neoplasms/etiology
9.
Mt Sinai J Med ; 79(3): 376-87, 2012.
Article En | MEDLINE | ID: mdl-22678861

Transplantation is the treatment of choice for end-stage kidney, heart, lung, and liver disease. Short-term outcomes in solid-organ transplantation are excellent, but long-term outcomes remain suboptimal. Advances in immune suppression and human leukocyte antigen matching techniques have reduced the acute rejection rate to <10%. Chronic allograft injury remains problematic and is in part immune-mediated. This injury is orchestrated by a complex adaptive and innate immune system that has evolved to protect the organism from infection, but, in the context of transplantation, could result in allograft rejection. Such chronic injury is partially mediated by anti-human leukocyte antigen antibodies. Severe rejections have largely been avoided by the development of tissue-typing techniques and crossmatch testing, which are discussed in detail. Further advances in the understanding of T- and B-cell immunology have led to the development of new immunomodulatory therapies directed at prolonging allograft survival, including those that decrease antibody production as well as those that remove antibodies from circulation. Further application of these immunomodulatory therapies has allowed expansion of the donor pool in some cases by permitting ABO-incompatible transplantation and transplantation in patients with preformed antibodies. Although vast improvements have been made in allograft survival, patients must remain on lifetime immunosuppression. Withdrawal of immunosuppression almost always ultimately leads to allograft rejection. The ultimate dream of transplant biologists is the induction of tolerance, where immune function remains intact but the allograft is not rejected in the face of withdrawn immunosuppression. This, however, has remained a significant challenge in human studies.


Graft Rejection/prevention & control , Health Knowledge, Attitudes, Practice , Organ Transplantation/methods , Adaptive Immunity , Allergy and Immunology , Antibodies/immunology , B-Lymphocytes , Graft Rejection/immunology , Humans , Immunity, Innate , Immunosuppressive Agents/therapeutic use , Organ Transplantation/adverse effects , T-Lymphocytes
10.
Clin Transplant ; 26(3): E261-8, 2012.
Article En | MEDLINE | ID: mdl-22686949

Limited data exist on the effect of intravenous immunoglobulin (IVIg) on anti-HLA antibodies as determined by solid-phase assays. We reviewed our experience treating sensitized wait-listed kidney transplant recipients with IVIg as a method for desensitization and report our results utilizing Luminex single antigen (LSA) bead assay to quantify antibody reactivity (MFI). Fifteen patients with a cPRA > 40% received 2 g/kg IVIg per month for four months or until transplanted. LSA testing was performed before and after IVIg. Median MFI for anti-class I antibodies fell in 11 (73%) and increased in 4 (27%) patients after IVIg. Similar significant changes in MFI for anti-class II antibodies were observed in 10 patients (66%). Administration of IVIg was associated with a modest decrease in reactivity to both class I and II HLA antigens (median MFI change 493 and 1110, respectively; p < 0.0001) but did not significantly alter mean cPRA (85% before IVIg vs. 80% after IVIg; p = 0.1). Our data suggest a smaller effect of IVIg on HLA antibody reactivity than previously described, leading us to question how best to measure the efficacy of a desensitization protocol in current practice.


HLA Antigens/immunology , Hypersensitivity/drug therapy , Immunoglobulins, Intravenous/administration & dosage , Immunologic Factors/administration & dosage , Isoantibodies/immunology , Kidney Transplantation/immunology , Adolescent , Adult , Aged , Child , Desensitization, Immunologic , Female , Follow-Up Studies , Graft Rejection/prevention & control , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Transplantation Conditioning , Waiting Lists
11.
J Immunol ; 187(2): 1023-30, 2011 Jul 15.
Article En | MEDLINE | ID: mdl-21677143

Chronic allograft vasculopathy (CAV) contributes to heart transplant failure, yet its pathogenesis is incompletely understood. Although cellular and humoral alloimmunity are accepted pathogenic mediators, animal models suggest that T cells and Abs reactive to graft-expressed autoantigens, including cardiac myosin (CM), could participate. To test the relationship between CAV and anti-CM autoimmunity in humans, we performed a cross-sectional study of 72 heart transplant recipients: 40 with CAV and 32 without. Sera from 65% of patients with CAV contained anti-CM Abs, whereas <10% contained Abs to other autoantigens (p < 0.05), and only 18% contained anti-HLA Abs (p < 0.05 versus anti-CM). In contrast, 13% of sera from patients without CAV contained anti-CM Abs (p < 0.05; odds ratio [OR], associating CAV with anti-CM Ab = 13, 95% confidence interval [CI] 3.79-44.6). Multivariable analysis confirmed the association to be independent of time posttransplant and the presence of anti-HLA Abs (OR = 28, 95% CI 5.77-133.56). PBMCs from patients with CAV responded more frequently to, and to a broader array of, CM-derived peptides than those without CAV (p = 0.01). Detection of either CM-peptide-reactive T cells or anti-CM Abs was highly and independently indicative of CAV (OR = 45, 95% CI 4.04-500.69). Our data suggest detection of anti-CM immunity could be used as a biomarker for outcome in heart transplantation recipients and support the need for further studies to assess whether anti-CM immunity is a pathogenic mediator of CAV.


Cardiac Myosins/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Graft Rejection/immunology , Graft Rejection/pathology , Heart Transplantation/immunology , Heart Transplantation/pathology , Aged , Amino Acid Sequence , Animals , Autoantibodies/biosynthesis , Biomarkers/blood , Chronic Disease , Complement C4b/immunology , Cross-Sectional Studies , Endothelium, Vascular/metabolism , Female , Graft Rejection/diagnosis , HLA Antigens/immunology , Humans , Macrophages/immunology , Macrophages/pathology , Male , Middle Aged , Molecular Sequence Data , Neutrophil Infiltration/immunology , Peptide Fragments/immunology , Pilot Projects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
12.
J Am Soc Nephrol ; 22(6): 1168-78, 2011 Jun.
Article En | MEDLINE | ID: mdl-21566057

Although T and B cell alloimmunity contribute to transplant injury, autoimmunity directed at kidney-expressed, non-HLA antigens may also participate. Because the specificity, prevalence, and importance of antibodies to non-HLA antigens in late allograft injury are poorly characterized, we used a protein microarray to compare antibody repertoires in pre- and post-transplant sera from several cohorts of patients with and without transplant glomerulopathy. Transplantation routinely induced changes in antibody repertoires, but we did not identify any de novo non-HLA antibodies common to patients with transplant glomerulopathy. The screening studies identified three reactivities present before transplantation that persisted after transplant and strongly associated with transplant glomerulopathy. ELISA confirmed that reactivity against peroxisomal-trans-2-enoyl-coA-reductase strongly associated with the development of transplant glomerulopathy in independent validation sets. In addition to providing insight into effects of transplantation on non-HLA antibody repertoires, these results suggest that pretransplant serum antibodies to peroxisomal-trans-2-enoyl-coA-reductase may predict prognosis in kidney transplantation.


Antibodies/blood , Kidney Glomerulus/pathology , Kidney Transplantation/pathology , Oxidoreductases Acting on CH-CH Group Donors/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies/immunology , Case-Control Studies , Child , Enzyme-Linked Immunosorbent Assay , Humans , Kidney Glomerulus/immunology , Kidney Transplantation/immunology , Middle Aged , Predictive Value of Tests , Prognosis , Protein Array Analysis , Young Adult
13.
Transplantation ; 88(10): 1157-8, 2009 Nov 27.
Article En | MEDLINE | ID: mdl-19935367

Chronic allograft injury remains the leading cause of graft loss despite improvements in immunosuppression, clinical risk stratification, and state-of-the-art antibody testing. Emerging results indicate that T-cell immune monitoring by cytokine enzyme-linked immunosorbent spot, as part of a comprehensive risk assessment platform, has the potential to guide decision making and improve outcomes after transplantation.


Graft Rejection/immunology , Kidney Transplantation/immunology , Monitoring, Immunologic/methods , T-Lymphocytes/immunology , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology , Cohort Studies , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay/methods , Graft Rejection/pathology , Humans , Interferon-gamma/immunology , Kidney Transplantation/pathology , Renal Dialysis , Risk Assessment , Treatment Failure , Treatment Outcome
14.
Clin Transplant ; 23(6): 951-7, 2009.
Article En | MEDLINE | ID: mdl-19624692

INTRODUCTION: The aim of this study is to investigate the prevalence, predictors, and clinical outcomes of acute antibody-mediated rejection (AAMR). METHODS: Retrospective analysis of 833 adult patients who received kidney transplantation between 1/1/2001 and 8/15/2007. RESULTS: The prevalence of AAMR and acute cellular rejection was 2% and 8.2%, respectively. Eight patients had type I, seven type II, and two type III AAMR. All patients had at least one strong donor-specific anti-HLA antibodies (DSA) with a median fluorescence intensity (MFI) value of over 6000 and the mean number of strong DSAs was 2.0 +/- 0.7. Fifteen of 17 patients received pre-transplant desensitization treatment. During a median 28 months of follow-up (range: 12-38 months), two patients died (88% patient survival), and nine lost their allografts (35% graft survival). While all type I AAMR patients responded to treatment, all type III patients, and four of seven patients with type II AAMR lost their allografts earlier, and three type II AAMR patients later due to transplant glomerulopathy. CONCLUSIONS: AAMR is mainly seen in patients with pre-transplant strong DSAs. There is a striking difference in clinical outcomes of AAMR that types II and III AAMR patients have poor prognosis compared to type I AAMR patients.


Graft Rejection/immunology , HLA Antigens/immunology , Isoantibodies/blood , Kidney Transplantation/immunology , Adult , Aged , Biopsy , Cross Reactions , Disease Progression , Female , Follow-Up Studies , Graft Rejection/drug therapy , Graft Rejection/pathology , Humans , Immunosuppressive Agents/therapeutic use , Kidney Failure, Chronic/surgery , Kidney Transplantation/pathology , Male , Middle Aged , Prognosis , Retrospective Studies , Severity of Illness Index , Time Factors
16.
Curr Opin Organ Transplant ; 13(4): 419-24, 2008 Aug.
Article En | MEDLINE | ID: mdl-18685339

PURPOSE OF REVIEW: Chronic injury and late allograft loss remain major causes of morbidity in clinical transplantation. Biomarkers that can reliably assess the risk of posttransplant complications are required to direct and individualize therapy aimed at prolonging graft survival and improving patient health. The purpose of this review is to provide a framework for understanding how to use biomarkers in the context of clinical transplantation and to summarize current data on available noninvasive cellular-based immune monitoring methods to predict transplant outcome. RECENT FINDINGS: New microarray and gene profiling data reveal peripheral blood cell gene expression patterns that identify operational tolerance, raising the possibility that the measurements can be used to direct immunosuppression withdrawal. Additional data support the use of selective urine gene products and soluble CD30 measurements in serum as reliable biomarkers of acute graft injury. Finally, recent studies demonstrate that measurement of T-cell alloimmunity by cytokine enzyme-linked immunospot is a promising, supplementary pretransplant risk assessment tool. SUMMARY: Recently published studies in organ transplantation suggest that results derived from assays focused on markers of T-cell immunity can segregate transplant candidates or recipients into high and low-risk subgroups for posttransplant graft injury. Larger prospective studies are needed, however, before any proposed biomarker can be incorporated into the transplant physicians' armamentarium to guide individualized therapeutic decision-making.


Biomarkers/analysis , Genetic Markers , Graft Rejection/diagnosis , Graft Survival , Organ Transplantation/adverse effects , T-Lymphocytes/immunology , Biological Assay , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling/methods , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/prevention & control , Humans , Immunosuppressive Agents/therapeutic use , Oligonucleotide Array Sequence Analysis , Patient Selection , Predictive Value of Tests , Risk Assessment , Transplantation Tolerance
17.
Clin J Am Soc Nephrol ; 3(4): 1160-7, 2008 Jul.
Article En | MEDLINE | ID: mdl-18337549

BACKGROUND AND OBJECTIVES: The objective of this study was to investigate the effects of desensitization protocols using intravenous Ig with or without plasmapheresis in patients with donor-specific anti-HLA antibodies on prevention of antibody-mediated rejection and downregulation of donor-specific antibodies. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Thirty-five complement-dependent cytotoxicity T cell cross-match-negative but complement-dependent cytotoxicity B cell and/or flow cytometry cross-match-positive kidney transplant recipients were treated with high-dosage intravenous Ig plus Thymoglobulin induction treatment. Donor-specific antibody strength was stratified as strong, medium, or weak by Luminex flow beads. Group 1 patients had weak/moderate and group 2 strong donor-specific antibodies RESULTS: Whereas no group 1 patients had acute rejection, 66% of group 2 had acute rejection (44% antibody-mediated rejection, 22% cellular rejection). The protocol was then changed to the addition of peritransplantation plasmapheresis to patients with strong donor-specific antibodies (group 3). This change resulted in a dramatic decrease in the acute rejection rate to 7%. During a median 18 mo of follow-up, patient survival was 100, 100, and 93% and graft survival was 100, 78, and 86% in groups 1, 2, and 3, respectively. During follow-up, 17 (52%) patients lost donor-specific antibodies completely, and 10 (30%) lost some of donor-specific antibodies and/or decreased the strength of existing donor-specific antibodies. CONCLUSIONS: These results indicated that in patients with strong donor-specific antibodies, the addition of plasmapheresis to high-dosage intravenous Ig decreases the incidence of acute rejection. The majority of the patients, whether they received intravenous Ig alone or with plasmapheresis, lost their donor-specific antibodies during follow-up.


Graft Rejection/prevention & control , HLA Antigens/immunology , Immunoglobulins, Intravenous/therapeutic use , Isoantibodies/blood , Kidney Transplantation/immunology , Plasmapheresis , Adult , Aged , Antibodies, Monoclonal/therapeutic use , Antilymphocyte Serum , Cross Reactions , Female , Flow Cytometry , Graft Rejection/epidemiology , Graft Rejection/immunology , Graft Rejection/mortality , Histocompatibility Testing , Humans , Immunoglobulins, Intravenous/adverse effects , Incidence , Kidney Transplantation/mortality , Male , Middle Aged , Plasmapheresis/adverse effects , Time Factors , Treatment Outcome
18.
Endocrinol Metab Clin North Am ; 36(4): 1039-49; x, 2007 Dec.
Article En | MEDLINE | ID: mdl-17983935

Kidney transplantation is the most preferred treatment for end-stage renal disease because it improves not only the patient's survival compared with dialysis, but also the quality of life. Preemptive transplantation is transplantation performed prior to the initiation of renal dialysis. Recent observational studies have shown increased patient and graft survival with preemptive transplantation, compared to patients receiving transplantation after the initiation of dialysis. Preemptive simultaneous pancreas and kidney transplantation in type 1 diabetic recipients has also been shown to improve patient survival. These results indicate the importance of early referral of patients who have chronic kidney disease to nephrologists and transplant centers.


Diabetic Nephropathies/surgery , Kidney Failure, Chronic/surgery , Kidney Transplantation , Humans , Pancreas Transplantation , Referral and Consultation , Renal Dialysis
19.
Clin J Am Soc Nephrol ; 2(6): 1261-7, 2007 Nov.
Article En | MEDLINE | ID: mdl-17942771

BACKGROUND AND OBJECTIVES: Transplant glomerulopathy (TGP) has been proposed to be a component of chronic antibody-mediated rejection (AMR). We have studied 36 patients with TGP and 51 patients with chronic allograft nephropathy (CAN) but without TGP for C4d staining and donor-specific anti-HLA antibodies (DSA) to investigate the alloantibody-mediated mechanisms. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Allograft biopsies were stained with C4d staining and DSAs were studied by Luminex Flow Beads. Allograft biopsies were done at a mean of 5.3 +/- 5.0 and 5.6 +/- 4.6 yr after transplantation in patients with CAN and TGP, respectively. RESULTS: The mean creatinine level at the time of the biopsy was 2.7 +/- 1.2 mg/dl in each group. Proteinuria of >1.0 g/d was more common in patients with TGP (61 versus 25%; P = 0.002). Whereas three patients with TGP had a history of acute AMR, none of the patients with CAN had. Mean chronicity score of the biopsies were 1.7 +/- 0.7 in patients with CAN and 1.9 +/- 0.8 in patients with TGP. Biopsies from only two (4%) patients with CAN and four (11%) patients with TGP had diffuse C4d positivity. DSA were found in 36% of TGP and 33% of CAN patients. CONCLUSIONS: These results suggest that a substantial number of patients with TGP did not have positive C4d staining or DSA, indicating that a non-alloantibody-mediated process may be involved in the development of TGP in some patients.


Complement C4b/analysis , Graft Rejection/etiology , HLA Antigens/immunology , Isoantibodies/blood , Kidney Glomerulus/pathology , Kidney Transplantation/adverse effects , Peptide Fragments/analysis , Tissue Donors , Adult , Aged , Chronic Disease , Female , Graft Rejection/immunology , Graft Rejection/pathology , Humans , Male , Middle Aged , Staining and Labeling , Transplantation, Homologous
20.
J Clin Hypertens (Greenwich) ; 6(9): 494-8; quiz 499-500, 2004 Sep.
Article En | MEDLINE | ID: mdl-15365276

Elevated plasma homocysteine (Hcy) concentration is considered a risk factor for cardiovascular disease and may also be associated with hypertension. Although links have been established between hyperhomocysteinemia and elevated risk for cardiovascular events, the precise role of plasma Hcy in cardiovascular disease is unclear. Plasma Hcy increases with aging and is associated with other health-related behaviors, including smoking and diet patterns. Both epidemiologic and longitudinal clinical investigations have investigated the possible contribution of plasma Hcy to cardiovascular disease, and most report an association of plasma Hcy with the risk for cardiovascular and cerebral events. Some reports describe a significant relationship between Hcy and blood pressure levels, as well as higher Hcy in hypertensives compared to normotensives. Other studies find that the effect of Hcy disappears following adjustment for other risk factors. Because Hcy cosegregates with other risk factors, it has been difficult to identify an independent effect of Hcy on cardiovascular disease or hypertension. Hcy can be modified to some extent by vitamin supplementation. Hcy reduction may have some benefit in reducing cardiovascular risk in some patients, particularly the elderly. Because the question of an independent role of Hcy on risk for cardiovascular disease has not been determined, the assessment and treatment of Hcy should be approached in the context of other modifiable risk factors.


Cardiovascular Diseases/blood , Homocysteine/blood , Hypertension/blood , Humans
...