Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Bone Marrow Transplant ; 52(10): 1355-1360, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28287636

ABSTRACT

Transplant-associated thrombotic microangiopathy (TA-TMA) is an early complication of hematopoietic cell transplantation (HCT). A high mortality rate is documented in patients who are refractory to calcineurin inhibitor cessation. Estimates of TA-TMA prevalence vary significantly and are higher in allogeneic compared with autologous HCT. Furthermore, our understanding of the pathophysiology that is strongly related to diagnosis and treatment options is limited. Recent evidence has linked TA-TMA with atypical hemolytic uremic syndrome, a disease of excessive activation of the alternative pathway of complement, opening the Pandora's box in treatment options. As conventional treatment management is highly inefficient, detection of complement activation may allow for early recognition of patients who will benefit from complement inhibition. Preliminary clinical results showing successful eculizumab administration in children and adults with TA-TMA need to be carefully evaluated. Therefore, realizing the unmet needs of better understanding TA-TMA in this complex setting, we aimed to summarize current knowledge focusing on (1) critical evaluation of diagnostic criteria, (2) epidemiology and prognosis, (3) recent evidence of complement activation and endothelial damage and (4) treatment options.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Complement Activation , Hematopoietic Stem Cell Transplantation , Thrombotic Microangiopathies , Allografts , Atypical Hemolytic Uremic Syndrome/etiology , Atypical Hemolytic Uremic Syndrome/metabolism , Atypical Hemolytic Uremic Syndrome/mortality , Atypical Hemolytic Uremic Syndrome/physiopathology , Autografts , Humans , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/metabolism , Thrombotic Microangiopathies/mortality , Thrombotic Microangiopathies/physiopathology
2.
Lupus ; 26(7): 773-776, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27687020

ABSTRACT

Systemic lupus erythematosus (SLE), a disorder of the immune system, is potentially curable by allogeneic bone marrow transplantation (alloBMT). Until recently, alloBMT was limited by donor availability and toxicity. Reduced intensity conditioning (RIC) combined with post-transplantation cyclophosphamide (PTCy) has improved the availability and safety of alloBMT permitting its exploration in severe-refractory autoimmune illnesses. We report the six-year follow-up of a young female whose refractory SLE-associated nephrosis resolved after RIC alloBMT with PTCy.


Subject(s)
Bone Marrow Transplantation/methods , Cyclophosphamide/administration & dosage , Lupus Erythematosus, Systemic/therapy , Transplantation Conditioning/methods , Adult , Bone Marrow Transplantation/adverse effects , Female , Follow-Up Studies , Humans , Immunosuppressive Agents/administration & dosage , Lupus Erythematosus, Systemic/physiopathology , Transplantation, Homologous , Treatment Outcome
5.
Ann Oncol ; 21(6): 1203-1210, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19880437

ABSTRACT

BACKGROUND: Rituximab may improve transplant outcomes but may delay immunologic recovery. PATIENTS AND METHODS: Seventy-seven patients with low-grade or mantle cell lymphoma received autologous stem-cell transplantation (ASCT) on a phase II study. Rituximab 375 mg/m(2) was administered 3 days before mobilization-dose cyclophosphamide, then weekly for four doses after count recovery from ASCT. Immune reconstitution was assessed. RESULTS: Sixty percent of transplants occurred in first remission. Actuarial event-free survival (EFS) and overall survival (OS) were 60% and 73%, respectively, at 5 years, with 7.2-year median follow-up for OS in surviving patients. Median EFS was 8.3 years. Older age and transformed lymphomas were independently associated with inferior EFS, whereas day 60 lymphocyte counts did not predict EFS or late infections. Early and late transplant-related mortality was 1% and 8%, with secondary leukemia in two patients. B-cell counts recovered by 1-2 years; however, the median IgG level remained low at 2 years. Late-onset idiopathic neutropenia, generally inconsequential, was noted in 43%. CONCLUSION: ASCT with rituximab can produce durable remissions on follow-up out to 10 years. Major infections do not appear to be significantly increased or to be predicted by immune monitoring.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Immune System/physiology , Lymphoma, Mantle-Cell , Lymphoma , Recovery of Function/immunology , Stem Cell Transplantation/methods , Adult , Aged , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/administration & dosage , Combined Modality Therapy , Drug Administration Schedule , Female , Humans , Immunotherapy , Lymphoma/immunology , Lymphoma/pathology , Lymphoma/rehabilitation , Lymphoma/therapy , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/rehabilitation , Lymphoma, Mantle-Cell/therapy , Male , Middle Aged , Neoplasm Staging , Rituximab , Transplantation Immunology , Transplantation, Autologous
7.
Bone Marrow Transplant ; 43(6): 477-80, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18978825

ABSTRACT

Graft failure after allogeneic blood or marrow transplantation, although generally uncommon, can be a devastating complication. This report includes the outcome of nine patients who received a salvage transplant for failure to engraft after one (n=8) or 2 (n=1) prior transplants. Eight patients received allografts from the original donor. All received fludarabine 30 mg/m(2) i.v. and alemtuzumab 20 mg i.v. daily from days -6 to -2. Daily CYA was begun on day -2, and the allograft was infused on day 0. The therapy was well tolerated with low toxicity, and all nine patients engrafted, recovering neutrophils at a median of 12 days after transplant. Four patients died: two of relapse, one of a fungal infection in the setting of GVHD and one of multiple sclerosis. The combination of fludarabine and alemtuzumab is an effective and well-tolerated salvage conditioning regimen for patients who experience graft failure after blood or marrow transplants.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neoplasm/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bone Marrow Transplantation/methods , Transplantation Conditioning/methods , Vidarabine/analogs & derivatives , Adult , Alemtuzumab , Antibodies, Monoclonal, Humanized , Female , Graft Rejection , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Recurrence , Retrospective Studies , Salvage Therapy/methods , Treatment Outcome , Vidarabine/administration & dosage
8.
Lupus ; 17(12): 1079-85, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19029275

ABSTRACT

In spite of current therapies, the overall health status of patients with SLE is poor. High-dose cyclophosphamide (50 mg/kg for 4 days) with or without stem-cell rescue has been introduced as a new therapy for severe SLE, including renal and central nervous system (CNS)-SLE. Long-term durable responses have been found to be 40%. A randomised clinical trial was completed comparing high-dose cyclophosphamide with monthly intravenous cyclophosphamide (750 mg/m squared bovine serum albumin) in patients with SLE who need cyclophosphamide for the first time. The primary outcome of the trial was complete clinical response. In this report, we compare the treatment groups with respect to quality of life. The patients in this study had a mean age of 35.3 +/- 10.1 years, were of Caucasian (35%), African-American (51%), Hispanic (8%) and Asian (6%) people, and 88% were women. The organ leading to treatment was renal lupus in 29%, CNS-lupus in 45% and other organs in 26%. Quality of life was measured at each visit using the Medical Outcome Study Short-Form 36 (SF-36). At 6 months, the patients in the high-dose cyclophosphamide trial arm had significantly greater improvement than patients in the monthly intravenous cyclophosphamide arm (P = 0.026; P = 0.0082, respectively) in the categories of general health and social functioning. At 18 months, the improvement in the role-physical score was significantly greater in the high-dose cyclophosphamide trial arm than in the monthly-dose cyclophosphamide arm (P = 0.025). At the end of the two and a half-year study, there were no significant differences between the groups with respect to changes in SF-36. By pooling the groups, at 30 months, there was a statistically significant (P < 0.05) improvement over baseline in 6 of the 8 SF-36 domains. This study shows earlier improvement in SF-36 measures at 6 months in the high-dose cyclophosphamide group but equal improvement in both arms at two and one and a half years. Eventual improvements in quality-of-life with both cyclophosphamide regimens are clinically meaningful to both patients and treating physicians.


Subject(s)
Cyclophosphamide/administration & dosage , Immunosuppressive Agents/administration & dosage , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/physiopathology , Quality of Life , Adult , Disability Evaluation , Dose-Response Relationship, Drug , Emotions , Female , Humans , Injections, Intravenous , Lupus Erythematosus, Systemic/psychology , Male , Middle Aged , Motor Activity , Severity of Illness Index
9.
Bone Marrow Transplant ; 42(8): 523-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18622413

ABSTRACT

Allogeneic blood or marrow transplantation (BMT) is potentially curative for a variety of life-threatening nonmalignant hematologic diseases such as paroxysmal nocturnal hemoglobinuria (PNH) and hemoglobinopathies. The application of BMT to treat these disorders is limited by the lack of suitable donors and often end-organ damage from the underlying disease. We treated three patients with thrombotic PNH, one of whom also had sickle cell disease, with a nonmyeloablative, HLA-haploidentical BMT with post-transplant CY. Rapid engraftment without GVHD occurred in two of the patients, including the patient with sickle cell disease. Both patients are disease free with full donor chimerism and require no immunosuppressive therapy, with follow-up of 1 and 4 years, respectively. Nonmyeloablative, HLA-haploidentical BMT with post-transplant CY is a promising approach for patients with life-threatening nonmalignant hematologic disease who lack an HLA-matched sibling donor.


Subject(s)
Bone Marrow Transplantation , Budd-Chiari Syndrome/therapy , Cyclophosphamide/administration & dosage , Graft Survival/drug effects , Immunosuppressive Agents/administration & dosage , Adult , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/therapy , Budd-Chiari Syndrome/complications , Female , Follow-Up Studies , Histocompatibility Testing , Humans , Male , Transplantation, Homologous
10.
Ann Rheum Dis ; 67(6): 775-81, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17974598

ABSTRACT

OBJECTIVE: To investigate the safety and tolerability of high-dose cyclophosphamide without stem cell rescue in scleroderma. METHODS: An open-label, single-site, uncontrolled study design entered patients with active diffuse cutaneous scleroderma. Patients were treated with cyclophosphamide (50 mg/kg) intravenously daily for 4 consecutive days (total 200 mg/kg) followed by granulocyte colony-stimulating factor (5 microg/kg/day). The primary clinical efficacy end point was the modified Rodnan skin score (mRSS). Secondary end points included the Health Assessment Questionnaire-Disability Index (HAQ-DI), physician global assessment (PGA) and pulmonary function tests. RESULTS: Six patients (4 men, 2 women) aged 19-60 years were entered into the study. One patients died early in the protocol, thus five patients had follow-up data. The percentage reduction of the mRSS in these five evaluable patients within 1 month of treatment was 60%, 55%, 41%, 31% and 0%. The patient with no decline in skin score at 1 month showed a decrease in skin score from 41 to 26 by the 3-month visit, a 37% improvement. Three patients sustained the improvement after treatment for 24, 12 and 12 months. Two patients relapsed at 12 and 6 months after treatment. The PGA and HAQ-DI scores improved in five of the six patients by 72% and 79% respectively at 3 months. The only serious adverse event was a death that occurred owing to infection after neutrophil count recovery. CONCLUSIONS: High-dose cyclophosphamide without stem cell rescue can lead to a clinically significant improvement in skin score and measures of disease severity in patients with diffuse cutaneous scleroderma.


Subject(s)
Cyclophosphamide/administration & dosage , Immunosuppressive Agents/administration & dosage , Scleroderma, Localized/drug therapy , Adult , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Drug Administration Schedule , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Injections, Intravenous , Male , Middle Aged , Scleroderma, Localized/pathology , Skin/pathology , Treatment Outcome
11.
J Thromb Haemost ; 4(5): 1114-24, 2006 May.
Article in English | MEDLINE | ID: mdl-16689766

ABSTRACT

BACKGROUND: Tissue factor pathway inhibitor (TFPI) lacks a membrane attachment signal but it remains associated with the endothelial surface via its association with an, as yet, unidentified glycosyl phosphatidylinositol (GPI)-anchored co-receptor. OBJECTIVES/METHODS: Cellular trafficking of TFPI within aerolysin-resistant ECV304 and EA.hy926 cells, which do not express GPI-anchored proteins on their surface, was compared with their wild-type counterparts. RESULTS AND CONCLUSIONS: Although aerolysin-resistant cells produce normal amounts of TFPI mRNA, TFPI is not expressed on the cell surface and total cellular TFPI is greatly decreased compared with wild-type cells. Additionally, normal, not increased, amounts of TFPI are secreted into conditioned media indicating that TFPI is degraded within the aerolysin-resistant cells. Confocal microscopy and studies using metabolic inhibitors demonstrate that aerolysin-resistant cells produce TFPI and transport it into the Golgi with subsequent degradation in lysosomes. The experimental results provide no evidence that cell surface TFPI originates from secreted TFPI that binds back to a GPI-anchored protein. Instead, the data suggest that TFPI tightly, but reversibly, binds to a GPI anchored co-receptor in the ER/Golgi. The co-receptor then acts as a molecular chaperone for TFPI by trafficking it to the cell surface of wild-type cells or to lysosomes of aerolysin-resistant cells. TFPI that escapes co-receptor binding is secreted through the same pathway in both wild-type and aerolysin-resistant cells. The data provide a framework for understanding how TFPI is expressed on endothelium.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Lipoproteins/metabolism , Bacterial Toxins/pharmacology , Biological Transport , Cell Line , Cell Membrane/metabolism , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Lipoproteins/genetics , Polymerase Chain Reaction , Pore Forming Cytotoxic Proteins , RNA, Messenger/genetics , Transcription, Genetic/drug effects
12.
Leukemia ; 20(3): 458-62, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16437138

ABSTRACT

Aplastic anemia (AA) and hypoplastic myelodysplastic syndromes (hMDS) are often difficult to distinguish. However, an accurate diagnosis is important because the prognosis and treatment of these diseases may differ. CD34+ hematopoietic progenitors are central to the pathogenesis of both disorders; they are the targets of the autoimmune attack in AA and neoplastic transformation in MDS. The aim of this study was to assess whether bone marrow CD34+ cell numbers could be used in differentiating between AA and hMDS. The percentage of bone marrow CD34+ cells was normal or increased (mean -3.5+0.5%, range 1-7%) in 15 of 35 patients studied, and low (mean -0.13 +/- 0.02%, range 0.02-0.36%) in 20 of 35 patients. All patients with a normal or increased percentage of CD34+ cells were ultimately diagnosed with hMDS based on the detection of clonal cytogenetic abnormalities or progression to refractory anemia with excess blasts/acute myeloid leukemia. All patients with low marrow CD34+ cell numbers met standard clinical criteria for AA and have not demonstrated neoplastic transformation with follow-up. Quantification of marrow CD34+ cells may serve as an important tool for distinguishing between AA and hMDS.


Subject(s)
Anemia, Aplastic/immunology , Antigens, CD34/immunology , Bone Marrow Cells/immunology , Myelodysplastic Syndromes/immunology , Adult , Humans , Prognosis , Retrospective Studies
13.
Leukemia ; 18(10): 1740-2, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15356647

ABSTRACT

Secondary myelodysplastic syndrome (MDS)/acute leukemia frequently evolves from severe aplastic anemia (SAA) following immunosuppressive therapy. Secondary clonal cytogenetic abnormalities have now been reported after noncytotoxic therapy in two additional settings: all trans retinoic acid (ATRA) treatment of acute promyelocytic leukemia (APL) and imatinib for chronic myeloid leukemia (CML). We propose that SAA, APL, CML, and MDS represent different manifestations of generalized insults to the bone marrow. In SAA, the insult to hematopoietic progenitors leads to an immune attack, while in APL, CML, and MDS, it gives rise to the malignant clones. A primary insult to bone marrow could simultaneously lead to several abnormal hematopoietic cell clones, with one dominating and the others present but below the level of detection. Such a 'field leukemogenic effect' would be analogous to the 'field cancerization effect' described in solid tumors. Nonspecific cytotoxic therapies, including antileukemic chemotherapy and allogeneic transplantation, have broad activity that could inhibit both the overt disease and other undetectable coexistent abnormal clones. In contrast, disease-specific targeted therapy such as immunosuppressive therapy in aplastic anemia, ATRA in APL, or imatinib in CML would have no activity against other abnormal clones, allowing them to expand and become detectable as the dominant clone declines.


Subject(s)
Anemia, Aplastic/pathology , Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Promyelocytic, Acute/pathology , Anemia, Aplastic/drug therapy , Anemia, Aplastic/genetics , Clone Cells/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics
14.
Bone Marrow Transplant ; 30(4): 255-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12203143

ABSTRACT

Shwachman-Diamond syndrome is a rare autosomal recessive disorder characterized by exocrine pancreatic dysfunction, metaphyseal dysostosis and bone marrow dysfunction with a predilection towards severe hematologic complications. Allogeneic bone marrow transplantation has been used as a therapeutic approach for SDS patients with serious hematologic abnormalities with mixed results. There is some concern that these patients may be more susceptible to early (<100 days) transplant-related complications than other transplant groups. We report a patient who received a matched allogeneic transplant without developing serious early transplant-related complications, but eventually died from relapse of his disease. Although experience is limited, a review of the reported cases suggests patients with SDS may be transplanted without significant short-term morbidity and mortality.


Subject(s)
Bone Marrow Transplantation , Exocrine Pancreatic Insufficiency/therapy , Myelodysplastic Syndromes/therapy , Adult , Anemia, Refractory, with Excess of Blasts , Exocrine Pancreatic Insufficiency/diagnosis , Fatal Outcome , Humans , Male , Myelodysplastic Syndromes/diagnosis , Pancytopenia , Syndrome , Transplantation, Homologous , Transplantation, Isogeneic
15.
J Clin Oncol ; 19(23): 4314-21, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731514

ABSTRACT

PURPOSE: To evaluate the long-term outcome after allogeneic (allo) and autologous (auto) blood or marrow transplantation (BMT) in patients with relapsed or refractory Hodgkin's lymphoma (HL). PATIENTS AND METHODS: We analyzed the outcome of 157 consecutive patients with relapsed or refractory HL, who underwent BMT between March 1985 and April 1998. Patients

Subject(s)
Blood Transfusion , Bone Marrow Transplantation , Hodgkin Disease/therapy , Adolescent , Adult , Baltimore , Child , Disease-Free Survival , Female , Graft vs Host Disease , Hodgkin Disease/mortality , Humans , Longitudinal Studies , Male , Middle Aged , Recurrence , Survival Analysis , Transplantation, Autologous , Transplantation, Homologous , Treatment Outcome
16.
Br J Haematol ; 115(2): 476-82, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11703352

ABSTRACT

Aplastic anaemia and paroxysmal nocturnal haemoglobinuria (PNH) are closely related disorders. In PNH, haematopoietic stem cells that harbour PIGA mutations give rise to blood elements that are unable to synthesize glycosylphosphatidylinositol (GPI) anchors. Because the GPI anchor is the receptor for the channel-forming protein aerolysin, PNH cells do not bind the toxin and are unaffected by concentrations that lyse normal cells. Exploiting these biological differences, we have developed two novel aerolysin-based assays to detect small populations of PNH cells. CD59 populations as small as 0.004% of total red cells could be detected when cells were pretreated with aerolysin to enrich the PNH population. All PNH patients displayed CD59-deficient erythrocytes, but no myelodysplastic syndrome (MDS) patient or control had detectable PNH cells before or after enrichment in aerolysin. Only one aplastic anaemia patient had detectable PNH red cells before exposure to aerolysin. However, 14 (61%) had detectable PNH cells after enrichment in aerolysin. The inactive fluorescent proaerolysin variant (FLAER) that binds the GPI anchors of a number of proteins on normal cells was used to detect a global GPI anchor deficit on granulocytes. Flow cytometry with FLAER showed that 12 out of 18 (67%) aplastic anaemia patients had FLAER-negative granulocytes, but none of the MDS patients or normal control subjects had GPI anchor-deficient cells. These studies demonstrate that aerolysin-based assays can reveal previously undetectable multilineage PNH cells in patients with untreated aplastic anaemia. Thus, clonality appears to be an early feature of aplastic anaemia.


Subject(s)
Anemia, Aplastic/complications , Glycosylphosphatidylinositols/deficiency , Hematopoiesis/physiology , Hemoglobinuria, Paroxysmal/complications , Adult , Aged , Anemia, Aplastic/blood , Bacterial Toxins , Erythrocytes/metabolism , Female , Flow Cytometry , Glycosylphosphatidylinositols/blood , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/diagnosis , Hemolysin Proteins , Humans , Male , Middle Aged , Pore Forming Cytotoxic Proteins
17.
Ann Intern Med ; 135(7): 477-83, 2001 Oct 02.
Article in English | MEDLINE | ID: mdl-11578150

ABSTRACT

BACKGROUND: Severe aplastic anemia is a life-threatening bone marrow failure disorder. High-dose cyclophosphamide therapy followed by allogeneic bone marrow transplantation cures the disease. However, it requires a suitable donor and carries the risk for graft-versus-host disease. A small pilot study demonstrated that high-dose cyclophosphamide therapy without bone marrow transplantation leads to durable, treatment-free complete remission. OBJECTIVE: To confirm the safety and efficacy of high-dose cyclophosphamide therapy alone in patients with severe aplastic anemia. DESIGN: Uncontrolled clinical trial. SETTING: Three tertiary care hospitals. PATIENTS: 19 patients with untreated severe aplastic anemia. INTERVENTION: Cyclophosphamide, 50 mg/kg of body weight per day for 4 consecutive days. MEASUREMENTS: Probability of response and overall survival were measured. Complete remission was defined as normal blood count for age and sex. Partial remission was defined as independence from transfusion and an absolute neutrophil count greater than 0.5 x 10(9) cells/L without growth factor support. Nonresponders were patients who remained transfusion dependent or died. Relapse was defined as no longer meeting criteria for partial or complete remission. RESULTS: The median time to an absolute neutrophil count of 0.5 x 10(9) cells/L was 49 days. The probability of survival was 84% (95% CI, 59% to 95%) at 24 months. The probability of achieving treatment-free remission was 73% (CI, 51% to 91%) at 24 months, and the probability of achieving complete remission was 65% (CI, 39% to 89%) at 50 months. No responding patients have had relapse or have developed secondary clonal disorders. CONCLUSIONS: High-dose cyclophosphamide therapy without bone marrow transplantation produces durable treatment-free remission in severe aplastic anemia. This approach deserves further study in patients with severe aplastic anemia who are not suitable candidates for allogeneic bone marrow transplantation.


Subject(s)
Anemia, Aplastic/drug therapy , Cyclophosphamide/administration & dosage , Immunosuppressive Agents/administration & dosage , Adolescent , Adult , Aged , Anemia, Aplastic/blood , Anemia, Aplastic/mortality , Anti-Bacterial Agents/therapeutic use , Antiemetics/therapeutic use , Cyclophosphamide/adverse effects , Drug Administration Schedule , Drug Therapy, Combination , Female , Hematopoiesis , Humans , Immunosuppressive Agents/adverse effects , Leukocyte Count , Male , Middle Aged , Neutrophils , Ondansetron/therapeutic use , Pilot Projects , Platelet Transfusion , Prospective Studies , Remission Induction , Survival Analysis
19.
Transplantation ; 71(3): 482-4, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11233915

ABSTRACT

BACKGROUND: Alloimmunization is a major problem for patients being considered for solid organ transplantation and in patients who require blood transfusion support. We previously demonstrated that high-dose cyclophosphamide (200 mg/kg) without hematopoietic stem cell transplantation leads to durable complete remissions in aplastic anemia and other autoimmune disorders. We now examine the ability of high-dose cyclophosphamide to eliminate alloreactivity. METHODS: IgG-specific antibodies to HLA class I were assayed using enzyme-linked immunosorbent assays in 18 consecutive patients with severe aplastic anemia before and after treatment with high-dose cyclophosphamide. RESULTS: Anti-HLA antibodies were detected before or shortly after therapy in 5 of the 18 patients studied. Complete remission of aplastic anemia was achieved in four of these five patients. High-dose cyclophosphamide markedly reduced anti-HLA antibody titers in these four patients; they were completely eradicated in three patients. Only one patient did not achieve significant reduction in the alloantibody titer after high-dose cyclophosphamide. CONCLUSIONS: High-dose cyclophosphamide without stem cell transplantation can eradicate HLA-specific alloantibody.


Subject(s)
Cyclophosphamide/administration & dosage , Isoantibodies/drug effects , Adult , Anemia, Aplastic/immunology , Anemia, Aplastic/therapy , Antibodies/blood , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , HLA Antigens/immunology , Humans , Immunoglobulin G/analysis , Male , Middle Aged
20.
Cancer Res ; 61(2): 654-8, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11212264

ABSTRACT

Phosphatidylinositol glycan-A (PIGA) is a gene that encodes an element required for the first step in glycosylphosphatidylinositol (GPI) anchor assembly. Because PIGA is X-located, a single mutation is sufficient to abolish cell surface GPI-anchored protein expression. In this study, we investigated whether mutation of the PIGA gene could be exploited to identify mutator (Mut) phenotypes in cancer. We examined eight Mut colon cancer lines and four non-Mut colon cancers as controls. In every case, flow cytometric analyses of cells sorted for low fluorescence after staining for GPI-linked CD59 and CD55 revealed negative peaks in the Mut lines but not in the controls. Single cell cloning of purged and sorted GPI-anchor- HCT116 cells and sequencing of the PIGA gene in each clone uniformly showed mutations. Pretreatment of the Mut lines with anti-CD55 or anti-CD59 antibodies and complement or with the GPI-anchor-reactive bacterial toxin aerolysin enriched for the GPI-anchor- populations. Expansion of purged GPI-anchor+ cells in the Mut lines and analyses using aerolysin in conjunction with flow cytometry yielded PIGA gene mutation frequencies of 10(-5) to 10(-4), values similar to the mutation frequencies of the hprt gene. This novel approach allows for the detection of as yet undescribed repair or replication defects and in addition to its considerably greater ease of use than existing techniques and in principle would not require the production of cell lines.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Membrane Proteins/deficiency , Neoplasms/pathology , Biomarkers , CD55 Antigens/analysis , CD55 Antigens/immunology , CD59 Antigens/analysis , CD59 Antigens/immunology , Complement System Proteins/immunology , Cytotoxicity, Immunologic , Flow Cytometry , Gene Frequency , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Phenotype , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...