Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Pediatr Transplant ; 23(7): e13561, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31483086

RESUMO

The frequency, indications, and outcomes for readmission following pediatric heart transplantation are poorly characterized. A better understanding of this phenomenon will help guide strategies to address the causes of readmission. Data from the Clinical Trials in Organ Transplantation for Children (CTOTC-04) multi-institutional collaborative study were utilized to determine incidence of, and risk factors for, hospital readmission within 30 days and 1 year from initial hospital discharge. Among 240 transplants at 8 centers, 227 subjects were discharged and had follow-up. 129 subjects (56.8%) were readmitted within one year; 71 had two or more readmissions. The 30-day and 1-year freedom from readmission were 70.5% (CI: 64.1%, 76.0%) and 42.2% (CI: 35.7%, 48.7%), respectively. The most common indications for readmissions were infection followed by rejection and fever without confirmed infection, accounting for 25.0%, 10.6%, and 6.2% of readmissions, respectively. Factors independently associated with increased risk of first readmission within 1 year (Cox proportional hazard model) were as follows: transplant in infancy (P = .05), longer transplant hospitalization (P = .04), lower UNOS urgency status (2/IB vs 1A) at transplant (P = .04), and Hispanic ethnicity (P = .05). Hospital readmission occurs frequently in the first year following discharge after heart transplantation with highest risk in the first 30 days. Infection is more common than rejection as cause for readmission, with death during readmission being rare. A number of patient factors are associated with higher risk of readmission. A fuller understanding of these risk factors may help tailor strategies to reduce unnecessary hospital readmission.


Assuntos
Transplante de Coração , Readmissão do Paciente/estatística & dados numéricos , Complicações Pós-Operatórias/epidemiologia , Adolescente , Criança , Pré-Escolar , Feminino , Seguimentos , Humanos , Incidência , Lactente , Estimativa de Kaplan-Meier , Masculino , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/terapia , Modelos de Riscos Proporcionais , Fatores de Risco
2.
J Heart Lung Transplant ; 38(9): 972-981, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31324444

RESUMO

BACKGROUND: Immunosuppression strategies have changed over time in pediatric heart transplantation. Thus, comorbidity profiles may have evolved. Clinical Trials in Organ Transplantation in Children-04 is a multicenter, prospective, cohort study assessing the impact of pre-transplant sensitization on outcomes after pediatric heart transplantation. This sub-study reports 1-year outcomes among recipients without pre-transplant donor-specific antibodies (DSAs). METHODS: We recruited consecutive candidates (<21 years) at 8 centers. Sensitization status was determined by a core laboratory. Immunosuppression was standardized as follows: Thymoglobulin induction with tacrolimus and/or mycophenolate mofetil maintenance. Steroids were not used beyond 1 week. Rejection surveillance was by serial biopsy. RESULTS: There were 240 transplants. Subjects for this sub-study (n = 186) were non-sensitized (n = 108) or had no DSAs (n = 78). Median age was 6 years, 48.4% were male, and 38.2% had congenital heart disease. Patient survival was 94.5% (95% confidence interval, 90.1-97.0%). Freedom from any type of rejection was 67.5%. Risk factors for rejection were older age at transplant and presence of non-DSAs pre-transplant. Freedom from infection requiring hospitalization/intravenous anti-microbials was 75.4%. Freedom from rehospitalization was 40.3%. New-onset diabetes mellitus and post-transplant lymphoproliferative disorder (PTLD) occurred in 1.6% and 1.1% of subjects, respectively. There was no decline in renal function over the first year. Corticosteroids were used in 14.5% at 1 year. CONCLUSIONS: Pediatric heart transplantation recipients without DSAs at transplant and managed with a steroid avoidance regimen have excellent short-term survival and a low risk of first-year diabetes mellitus and PTLD. Rehospitalization remains common. These contemporary observations allow for improved caregiver and/or patient counseling and provide the necessary outcomes data to help design future randomized controlled trials.


Assuntos
Soro Antilinfocitário/uso terapêutico , Transplante de Coração , Imunossupressores/uso terapêutico , Ácido Micofenólico/uso terapêutico , Tacrolimo/uso terapêutico , Adolescente , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Glucocorticoides , Humanos , Lactente , Masculino , Medição de Risco , Fatores de Tempo , Resultado do Tratamento
3.
Am J Transplant ; 19(8): 2262-2273, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30920136

RESUMO

Genetic variation across the human leukocyte antigen loci is known to influence renal-transplant outcome. However, the impact of genetic variation beyond the human leukocyte antigen loci is less clear. We tested the association of common genetic variation and clinical characteristics, from both the donor and recipient, with posttransplant eGFR at different time-points, out to 5 years posttransplantation. We conducted GWAS meta-analyses across 10 844 donors and recipients from five European ancestry cohorts. We also analyzed the impact of polygenic risk scores (PRS), calculated using genetic variants associated with nontransplant eGFR, on posttransplant eGFR. PRS calculated using the recipient genotype alone, as well as combined donor and recipient genotypes were significantly associated with eGFR at 1-year posttransplant. Thirty-two percent of the variability in eGFR at 1-year posttransplant was explained by our model containing clinical covariates (including weights for death/graft-failure), principal components and combined donor-recipient PRS, with 0.3% contributed by the PRS. No individual genetic variant was significantly associated with eGFR posttransplant in the GWAS. This is the first study to examine PRS, composed of variants that impact kidney function in the general population, in a posttransplant context. Despite PRS being a significant predictor of eGFR posttransplant, the effect size of common genetic factors is limited compared to clinical variables.


Assuntos
Marcadores Genéticos , Variação Genética , Rejeição de Enxerto/diagnóstico , Transplante de Rim/efeitos adversos , Rim/fisiopatologia , Complicações Pós-Operatórias/diagnóstico , Medição de Risco/métodos , Adulto , Europa (Continente)/epidemiologia , Feminino , Seguimentos , Estudo de Associação Genômica Ampla , Taxa de Filtração Glomerular , Rejeição de Enxerto/epidemiologia , Rejeição de Enxerto/genética , Sobrevivência de Enxerto , Humanos , Falência Renal Crônica/genética , Falência Renal Crônica/cirurgia , Testes de Função Renal , Doadores Vivos/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/genética , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Transplantados/estatística & dados numéricos
4.
Am J Transplant ; 19(1): 98-109, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29985559

RESUMO

Noninvasive biomarkers are needed to monitor stable patients after kidney transplant (KT), because subclinical acute rejection (subAR), currently detectable only with surveillance biopsies, can lead to chronic rejection and graft loss. We conducted a multicenter study to develop a blood-based molecular biomarker for subAR using peripheral blood paired with surveillance biopsies and strict clinical phenotyping algorithms for discovery and validation. At a predefined threshold, 72% to 75% of KT recipients achieved a negative biomarker test correlating with the absence of subAR (negative predictive value: 78%-88%), while a positive test was obtained in 25% to 28% correlating with the presence of subAR (positive predictive value: 47%-61%). The clinical phenotype and biomarker independently and statistically correlated with a composite clinical endpoint (renal function, biopsy-proved acute rejection, ≥grade 2 interstitial fibrosis, and tubular atrophy), as well as with de novo donor-specific antibodies. We also found that <50% showed histologic improvement of subAR on follow-up biopsies despite treatment and that the biomarker could predict this outcome. Our data suggest that a blood-based biomarker that reduces the need for the indiscriminate use of invasive surveillance biopsies and that correlates with transplant outcomes could be used to monitor KT recipients with stable renal function, including after treatment for subAR, potentially improving KT outcomes.


Assuntos
Biomarcadores/sangue , Rejeição de Enxerto/diagnóstico , Transplante de Rim , Adulto , Idoso , Algoritmos , Biópsia , Feminino , Fibrose/diagnóstico , Taxa de Filtração Glomerular , Rejeição de Enxerto/sangue , Sobrevivência de Enxerto , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Valor Preditivo dos Testes , Resultado do Tratamento , Adulto Jovem
5.
Am J Transplant ; 18(9): 2135-2147, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29446208

RESUMO

Anti-HLA donor-specific antibodies are associated with worse outcomes after organ transplantation. Among sensitized pediatric heart candidates, requirement for negative donor-specific cytotoxicity crossmatch increases wait times and mortality. However, transplantation with positive crossmatch may increase posttransplantation morbidity and mortality. We address this clinical challenge in a prospective, multicenter, observational cohort study of children listed for heart transplantation (Clinical Trials in Organ Transplantation in Children-04 [CTOTC-04]). Outcomes were compared among sensitized recipients who underwent transplantation with positive crossmatch, nonsensitized recipients, and sensitized recipients without positive crossmatch. Positive crossmatch recipients received antibody removal and augmented immunosuppression, while other recipients received standard immunosuppression with corticosteroid avoidance. This first CTOTC-04 report summarizes study rationale and design and relates pretransplantation sensitization status using solid-phase technology. Risk factors for sensitization were explored. Of 317 screened patients, 290 were enrolled and 240 underwent transplantation. Core laboratory evaluation demonstrated that more than half of patients were anti-HLA sensitized. Greater than 80% of sensitized patients had class I (with or without class II) HLA antibodies, and one-third of sensitized patients had at least 1 HLA antibody with median fluorescence intensity of ≥8000. Logistic regression models demonstrated male sex, weight, congenital heart disease history, prior allograft, and ventricular assist device are independent risk factors for sensitization.


Assuntos
Antígenos HLA/imunologia , Transplante de Coração/métodos , Isoanticorpos/imunologia , Projetos de Pesquisa , Doadores de Tecidos , Tolerância ao Transplante/imunologia , Adolescente , Criança , Pré-Escolar , Feminino , Seguimentos , Teste de Histocompatibilidade , Humanos , Terapia de Imunossupressão , Lactente , Recém-Nascido , Isoanticorpos/sangue , Masculino , Prognóstico , Estudos Prospectivos , Fatores de Risco , Transplante Homólogo
6.
Transpl Int ; 30(7): 689-694, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28346714

RESUMO

Vaccine immunoprotection for Streptococcus pneumoniae is mediated by opsonizing antibodies targeting serotype-specific capsular polysaccharides. Quantitative antibody levels enzyme-linked immunosorbent assay (ELISA) and antibody-mediated opsonophagocytic assays (OPA) measure vaccine-induced protection; correlation of these assays in transplantation requires investigation. This study examines the laboratory assessment of antibody titers in vaccinated renal recipients. Streptococcus pneumoniae 19A is common in immunocompromised hosts and is represented in protein-conjugate vaccines (PCV) and polysaccharide vaccines (PSV). Antibodies to 19A in serial sera from 30 vaccinated renal transplant recipients were compared using ELISA and OPA assays. Subject titers were classified as protected or not by ELISA (>0.35 µg/ml) and OPA titer (>1:8). Antibody titers analyzed using McNemar's test indicate that protection measured by the two assays are not the same (P = 0.0078); simple linear regression of within-subject geometric means of 19A enzyme-linked immunosorbent assay (ELISA) antibody levels versus 19A opsonophagocytic assays (OPA) titers demonstrates significant correlation between the two assays (P < 0.001). Vaccination is increasingly important given increasing antimicrobial resistance worldwide. OPA and ELISA antibody assays do not correlate well using current values for protective immunity against the Pneumococcus in immunosuppressed transplant recipients. Future studies of vaccination in transplant recipients should evaluate protective antibody levels using both functional antibody assays and standard ELISA antibody titers. (ClinicalTrials.gov: NCT00307125).


Assuntos
Transplante de Rim/efeitos adversos , Infecções Pneumocócicas/diagnóstico , Infecções Pneumocócicas/etiologia , Testes Sorológicos/métodos , Streptococcus pneumoniae/isolamento & purificação , Adulto , Anticorpos Antibacterianos/sangue , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Imunoensaio/métodos , Hospedeiro Imunocomprometido , Infecções Pneumocócicas/prevenção & controle , Vacinas Pneumocócicas/uso terapêutico , Estudos Prospectivos , Streptococcus pneumoniae/imunologia , Transplantados
7.
Clin Exp Rheumatol ; 34(2 Suppl 96): S89-98, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27157394

RESUMO

OBJECTIVES: Fibromyalgia (FM) is a common pain disorder characterized by nociceptive dysregulation. The basic biology of FM is poorly understood. Herein we have used agnostic gene expression as a potential probe for informing its underlying biology and the development of a proof-of-concept diagnostic gene expression signature. METHODS: We analyzed RNA expression in 70 FM patients and 70 healthy controls. The isolated RNA was amplified and hybridized to Affymetrix® Human Gene 1.1 ST Peg arrays. The data was analyzed using Partek Genomics Suite version 6.6. RESULTS: Fibromyalgia patients exhibited a differential expression of 421 genes (p<0.001), several relevant to pathways for pain processing, such as glutamine/glutamate signaling and axonal development. There was also an upregulation of several inflammatory pathways and downregulation of pathways related to hypersensitivity and allergy. Using rigorous diagnostic modeling strategies, we show "locked" gene signatures discovered on Training and Test cohorts, that have a mean Area Under the Curve (AUC) of 0.81 on randomized, independent external data cohorts. Lastly, we identified a subset of 10 probesets that provided a diagnostic sensitivity for FM of 95% and a specificity of 96%. We also show that the signatures for FM were very specific to FM rather than common FM comorbidities. CONCLUSIONS: These findings provide new insights relevant to the pathogenesis of FM, and provide several testable hypotheses that warrant further exploration and also establish the foundation for a first blood-based molecular signature in FM that needs to be validated in larger cohorts of patients.


Assuntos
Fibromialgia/genética , Perfilação da Expressão Gênica , Transcriptoma/fisiologia , Adulto , Carboxipeptidases A/genética , Feminino , Fibromialgia/sangue , Fator de Transcrição GATA2/genética , Estudo de Associação Genômica Ampla , Humanos , Pessoa de Meia-Idade , Nociceptividade/fisiologia , Receptores de IgE/genética , Transdução de Sinais/genética
8.
J Am Soc Nephrol ; 27(2): 626-36, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26047788

RESUMO

Noninvasive diagnosis and prognostication of acute cellular rejection in the kidney allograft may help realize the full benefits of kidney transplantation. To investigate whether urine metabolites predict kidney allograft status, we determined levels of 749 metabolites in 1516 urine samples from 241 kidney graft recipients enrolled in the prospective multicenter Clinical Trials in Organ Transplantation-04 study. A metabolite signature of the ratio of 3-sialyllactose to xanthosine in biopsy specimen-matched urine supernatants best discriminated acute cellular rejection biopsy specimens from specimens without rejection. For clinical application, we developed a high-throughput mass spectrometry-based assay that enabled absolute and rapid quantification of the 3-sialyllactose-to-xanthosine ratio in urine samples. A composite signature of ratios of 3-sialyllactose to xanthosine and quinolinate to X-16397 and our previously reported urinary cell mRNA signature of 18S ribosomal RNA, CD3ε mRNA, and interferon-inducible protein-10 mRNA outperformed the metabolite signatures and the mRNA signature. The area under the receiver operating characteristics curve for the composite metabolite-mRNA signature was 0.93, and the signature was diagnostic of acute cellular rejection with a specificity of 84% and a sensitivity of 90%. The composite signature, developed using solely biopsy specimen-matched urine samples, predicted future acute cellular rejection when applied to pristine samples taken days to weeks before biopsy. We conclude that metabolite profiling of urine offers a noninvasive means of diagnosing and prognosticating acute cellular rejection in the human kidney allograft, and that the combined metabolite and mRNA signature is diagnostic and prognostic of acute cellular rejection with very high accuracy.


Assuntos
Aloenxertos/metabolismo , Rejeição de Enxerto/urina , Transplante de Rim , Rim/metabolismo , Doença Aguda , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Rejeição de Enxerto/metabolismo , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Prospectivos , Adulto Jovem
9.
N Engl J Med ; 369(1): 20-31, 2013 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-23822777

RESUMO

BACKGROUND: The standard test for the diagnosis of acute rejection in kidney transplants is the renal biopsy. Noninvasive tests would be preferable. METHODS: We prospectively collected 4300 urine specimens from 485 kidney-graft recipients from day 3 through month 12 after transplantation. Messenger RNA (mRNA) levels were measured in urinary cells and correlated with allograft-rejection status with the use of logistic regression. RESULTS: A three-gene signature of 18S ribosomal (rRNA)-normalized measures of CD3ε mRNA and interferon-inducible protein 10 (IP-10) mRNA, and 18S rRNA discriminated between biopsy specimens showing acute cellular rejection and those not showing rejection (area under the curve [AUC], 0.85; 95% confidence interval [CI], 0.78 to 0.91; P<0.001 by receiver-operating-characteristic curve analysis). The cross-validation estimate of the AUC was 0.83 by bootstrap resampling, and the Hosmer-Lemeshow test indicated good fit (P=0.77). In an external-validation data set, the AUC was 0.74 (95% CI, 0.61 to 0.86; P<0.001) and did not differ significantly from the AUC in our primary data set (P=0.13). The signature distinguished acute cellular rejection from acute antibody-mediated rejection and borderline rejection (AUC, 0.78; 95% CI, 0.68 to 0.89; P<0.001). It also distinguished patients who received anti-interleukin-2 receptor antibodies from those who received T-cell-depleting antibodies (P<0.001) and was diagnostic of acute cellular rejection in both groups. Urinary tract infection did not affect the signature (P=0.69). The average trajectory of the signature in repeated urine samples remained below the diagnostic threshold for acute cellular rejection in the group of patients with no rejection, but in the group with rejection, there was a sharp rise during the weeks before the biopsy showing rejection (P<0.001). CONCLUSIONS: A molecular signature of CD3ε mRNA, IP-10 mRNA, and 18S rRNA levels in urinary cells appears to be diagnostic and prognostic of acute cellular rejection in kidney allografts. (Funded by the National Institutes of Health and others.).


Assuntos
Quimiocina CXCL10/genética , Rejeição de Enxerto/diagnóstico , Peptídeos e Proteínas de Sinalização Intracelular/genética , Transplante de Rim , RNA Mensageiro/urina , RNA Ribossômico/urina , Doença Aguda , Adulto , Área Sob a Curva , Quimiocina CXCL10/urina , Feminino , Rejeição de Enxerto/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/urina , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , RNA Polimerase I , RNA Ribossômico 18S/urina , Curva ROC , Reação em Cadeia da Polimerase em Tempo Real , Sensibilidade e Especificidade , Transcriptoma
10.
Cancer Res ; 65(2): 622-31, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15695407

RESUMO

Antibody fragments are recognized as promising vehicles for delivery of imaging and therapeutic agents to tumor sites in vivo. The serum persistence of IgG1 and fragments with intact Fc region is controlled by the protective neonatal Fc receptor (FcRn) receptor. To modulate the half-life of engineered antibodies, we have mutated the Fc-FcRn binding site of chimeric anti-carcinoembryonic antigen (CEA) antibodies produced in a single-chain Fv-Fc format. The anti-CEA T84.66 single-chain Fv-Fc format wild-type and five mutants (I253A, H310A, H435Q, H435R, and H310A/H435Q, Kabat numbering system) expressed well in mammalian cell culture. After purification and characterization, effective in vitro antigen binding was shown by competition ELISA. Biodistribution studies in BALB/c mice using (125)I- and (131)I-labeled fragments revealed blood clearance rates from slowest to fastest as follows: wild-type > H435R > H435Q > I253A > H310A > H310A/H435Q. The terminal half-lives of the mutants ranged from 83.4 to 7.96 hours, whereas that of the wild-type was approximately 12 days. Additionally, (124)I-labeled wild-type, H435Q, I253A, H310A, and H310A/H435Q variants were evaluated in LS174T xenografted athymic mice by small animal positron emission tomography imaging, revealing localization to the CEA-positive xenografts. The slow clearing wild-type and H435Q constructs required longer to localize to the tumor and clear from the circulation. The I253A and H310A fragments showed intermediate behavior, whereas the H310A/H435Q variant quickly localized to the tumor site, rapidly cleared from the animal circulation and produced clear images. Thus, attenuating the Fc-FcRn interaction provides a way of controlling the antibody fragment serum half-life without compromising expression and tumor targeting.


Assuntos
Antígeno Carcinoembrionário/imunologia , Imunoconjugados/farmacocinética , Fragmentos de Imunoglobulinas/metabolismo , Radioisótopos do Iodo/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Animais , Antígeno Carcinoembrionário/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Fragmentos de Imunoglobulinas/biossíntese , Fragmentos de Imunoglobulinas/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mieloma Múltiplo/metabolismo , Tomografia por Emissão de Pósitrons , Distribuição Tecidual , Transplante Heterólogo
11.
Clin Cancer Res ; 10(15): 5014-21, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15297402

RESUMO

PURPOSE: The chimeric T84.66 (cT84.66) minibody is a novel engineered antibody construct (V(L)-linker-V(H)-C(H)3; 80 kDa) that demonstrates bivalent and high affinity (4 x 10(10) m(-1)) binding to carcinoembryonic antigen (CEA). The variable regions (V(L) and V(H)) assemble to form the antigen-combining sites, and the protein forms dimers through self-association of the C(H)3 domains. In animal models, the minibody demonstrated high tumor uptake, approaching that of some intact antibodies, substantially faster clearance than intact chimeric T84.66, and superior tumor-to-blood ratios compared with the cT84.66 F(ab')(2) fragment, making it attractive for further evaluation as an imaging and therapy agent. The purpose of this pilot clinical study was to determine whether (123)I-cT84.66 minibody demonstrated tumor targeting and was well tolerated as well as to begin to evaluate its biodistribution, pharmacokinetics, and immunogenicity in patients with colorectal cancer. EXPERIMENTAL DESIGN: Ten patients with biopsy-proven colorectal cancer (6 newly diagnosed, 1 pelvic recurrence, 3 limited metastatic disease) were entered on this study. Each received 5-10 mCi (1 mg) of (123)I-labeled minibody i.v. followed by serial nuclear scans and blood and urine sampling over the next 48-72 h. Surgery was performed immediately after the last nuclear scan. RESULTS: Tumor imaging was observed with (123)I-labeled minibody in seven of the eight patients who did not receive neoadjuvant therapy before surgery. Two patients received neoadjuvant radiation and chemotherapy, which significantly reduced tumor size before surgery and minibody infusion. At surgery, no tumor was detected in one patient and only a 2-mm focus was seen in the second patient. (123)I-labeled minibody tumor targeting was not seen in either of these pretreated patients. Mean serum residence time of the minibody was 29.8 h (range, 10.9-65.4 h). No drug-related adverse reactions were observed. All 10 patients were evaluated for immune responses to the minibody, with no significant responses observed. CONCLUSION: This pilot study represents one of the first clinical efforts to evaluate an engineered intermediate-molecular-mass radiolabeled antibody construct directed against CEA. cT84.66 minibody demonstrates tumor targeting to colorectal cancer and a faster clearance in comparison with intact antibodies, making it appropriate for further evaluation as an imaging and therapy agent. The mean residence time of the minibody in patients is longer than predicted from murine models. We therefore plan to further evaluate its biodistribution and pharmacokinetic properties with minibody labeled with a longer-lived radionuclide, such as (111)In.


Assuntos
Anticorpos/química , Anticorpos/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/terapia , Fragmentos de Imunoglobulinas/uso terapêutico , Imunoterapia/métodos , Radioisótopos do Iodo/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Cromatografia Líquida de Alta Pressão , Dimerização , Feminino , Humanos , Cinética , Masculino , Camundongos , Pessoa de Meia-Idade , Metástase Neoplásica , Projetos Piloto , Estrutura Terciária de Proteína , Radiometria , Fatores de Tempo , Tomografia Computadorizada de Emissão de Fóton Único
12.
Clin Cancer Res ; 9(16 Pt 1): 5842-52, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14676105

RESUMO

PURPOSE: Targeted systemic radiation therapy using radiolabeled antibodies results in tumor doses sufficient to produce significant objective responses in the radiosensitive hematological malignancies. Although comparable doses to tumor are achieved with radioimmunotherapy (RIT) in solid tumors, results have been modest primarily because of their relative lack of radiosensitivity. For solid tumors, as with external beam radiotherapy, RIT should have a more important clinical role if combined with other systemic, potentially radiation-enhancing chemotherapy agents and if used as consolidative therapy in the minimal tumor burden setting. The primary objective of this trial was to evaluate the feasibility and toxicities of systemic 90Y-chimeric T84.66 (cT84.66) anti-carcinoembryonic antigen RIT in combination with continuous infusion 5-fluorouracil (5-FU). EXPERIMENTAL DESIGN: Patients with chemotherapy-refractory metastatic colorectal cancer were entered. The study was designed for each patient to receive 90Y-cT84.66 anti-carcinoembryonic antigen at 16.6 mCi/m2 as an i.v. bolus infusion combined with 5-FU delivered as a 5-day continuous infusion initiated 4 h before antibody infusion. Cohorts of patients were entered at 5-FU dose levels of 700, 800, 900, and 1000 mg/m2/day. Upon reaching the highest planned dose level of 5-FU, a final cohort received 90Y-cT84.66 at 20.6 mCi/m2 and 5-FU at 1000 mg/m2/day. For all patients, Ca-diethylenetriaminepentaacetic acid at 125 mg/m2 every 12 h was administered for the first 72 h after 90Y-cT84.66. Patients were eligible to receive up to three cycles of 90Y-cT84.66/5-FU every 6 weeks. RESULTS: Twenty-one patients were treated on this study. All had been heavily pretreated with 19 having previously received 5-FU and 16 having failed two to four chemotherapy regimens. A maximum-tolerated dose of 16.6 mCi/m2 90Y-cT84.66 combined with 1000 mg/m2/day 5-FU was reached. These dose levels are comparable with maximum-tolerated dose levels of each agent alone. Thirteen patients received one cycle and 8 patients two cycles of therapy. Hematopoietic toxicity was dose-limiting and reversible. RIT did not appear to increase nonhematopoietic toxicities associated with 5-FU. Two of 19 patients assayed developed a human anti-chimeric antibody immune response after the first cycle of therapy, which is significantly less than that observed in a previous trial evaluating 90Y-cT84.66 alone. No objective responses were observed. However, 11 patients with progressive disease entering the study demonstrated radiological stable disease of 3-8 months duration and 1 patient demonstrated a mixed response. CONCLUSIONS: Results from this trial are encouraging and demonstrate the feasibility and possible advantages of combining continuous infusion 5-FU with 90Y-cT84.66 RIT. The addition of 5-FU does not appear to significantly enhance hematological toxicities of the radiolabeled antibody. In addition, 5-FU reduces the development of human anti-chimeric antibody response, permitting multicycle therapy in a larger number of patients. Future efforts should continue to focus on integrating radiation therapy delivered by radiolabeled antibodies into established 5-FU regimens.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/terapia , Fluoruracila/uso terapêutico , Radioimunoterapia , Proteínas Recombinantes de Fusão/uso terapêutico , Radioisótopos de Ítrio/uso terapêutico , Antimetabólitos Antineoplásicos/uso terapêutico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/secundário , Terapia Combinada , Estudos de Viabilidade , Humanos , Dose Máxima Tolerável
13.
Cancer Res ; 63(12): 3435-43, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12810682

RESUMO

Expression of the adenoviral E1A oncogene induces susceptibility of neoplastic cells from different species to both immune-mediated and chemotherapy-induced cell death. These effects of E1A are easily measured in vitro using cytotoxicity assays. However, conventional in vivo assays of tumor development lack similar precision for measurement of oncogene-induced changes in tumor cell traits. E1A expression in p53 mutant human breast carcinoma cells sensitized them in vitro to diverse immunological injuries and apoptosis triggered by chemotherapeutic agents, as predicted from studies of rodent tumor cells. Nude mice, which possess innate cellular immune defenses against E1A-expressing tumor cells, were used in a quantitative tumor induction assay to test the in vivo correlations of E1A-induced immunosensitivity and chemosensitivity of human tumor cells. Two distinct, E1A-induced breast cancer cell traits could be measured in nude mice: (a) increased tumor latency and (b) reduced efficiency of tumor induction. These results were confirmed in studies of E1A-expressing human fibrosarcoma cells. The results demonstrate that E1A-induced conversion of human cells from a cytolytic resistant to a cytolytic susceptible phenotype, as detected in vitro, translates into reduced tumorigenicity of cells confronted with innate immune defenses and exposed to chemotherapeutic agents in nude mice. However, the data also show that E1A expression does not completely eliminate the tumorigenicity of either established human tumor cells or of cells immortalized by E1A. This experimental approach should be useful for studies of the effects of other oncogene-related tumor cell traits on tumorigenicity and could be used for preclinical studies of different treatment strategies for human tumors.


Assuntos
Proteínas E1A de Adenovirus/fisiologia , Apoptose/fisiologia , Neoplasias Ósseas/patologia , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Osteossarcoma/patologia , Adenovírus Humanos/genética , Adenovírus Humanos/fisiologia , Animais , Antineoplásicos/uso terapêutico , Neoplasias Ósseas/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/imunologia , Linhagem Celular/transplante , Citotoxicidade Imunológica , Etoposídeo/uso terapêutico , Feminino , Fibrossarcoma/imunologia , Fibrossarcoma/patologia , Genes p53 , Humanos , Vigilância Imunológica , Rim , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Nus , Oncogenes , Osteossarcoma/imunologia , Ratos , Ratos Nus , Fatores de Tempo , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/imunologia , Proteína Supressora de Tumor p53/deficiência , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Am J Clin Oncol ; 26(1): 16-21, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12576918

RESUMO

For periampullary cancer,intraoperative radiation therapy (IORT) administered to the site with the highest locoregional recurrence risk carries the rationale to improve tumor control. An IORT effect on survival remains unclear. IORT impact on postoperative outcomes after pancreatectomy for adenocarcinoma was analyzed, with a specific attempt to correct for the nonrandom IORT treatment assignment, and to account for treatment group imbalances in the interpretation of outcome differences. A propensity-score-adjusted analysis, based on variable selection by logistic regression, was used to rebalance treatments. Between 1989 and 1999, 61 patients underwent partial or total pancreatectomy for a primary periampullary adenocarcinoma at the City of Hope National Medical Center. Diagnoses included pancreatic (n = 36), duodenal (n = 11), ampullary (n = 10), and bile duct cancer (n = 4). Thirty patients received IORT to the resection area, with a median dose of 15 Gy (range: 10-20), followed by postoperative external beam radiation (n = 24). Mortality was 0%, the complication rate 61%. Of 33 patients with a documented recurrence, 6 had an isolated locoregional recurrence only (1 IORT versus 5 no IORT, = 0.05); the systemic recurrence pattern differed as well (IORT 94%, no IORT 67%; = 0.04). IORT had no significant impact on hospital stay (overall median: 17 days), disease-free survival (16 months), and overall survival (23 months) when adjusted for those most relevant variables reflecting IORT treatment group assignment propensity. After adjustment for relevant propensity factors, IORT was not linked to a significantly increased risk for complications, hospital stay, or survival hazard. The recurrence pattern may be affected in some patients, but systemic recurrences predominate. We continue to explore IORT in combination with systemic chemotherapy.


Assuntos
Adenocarcinoma/radioterapia , Ampola Hepatopancreática , Neoplasias dos Ductos Biliares/radioterapia , Neoplasias Duodenais/radioterapia , Neoplasias Pancreáticas/radioterapia , Pancreaticoduodenectomia , Adenocarcinoma/cirurgia , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias dos Ductos Biliares/cirurgia , Neoplasias Duodenais/cirurgia , Feminino , Humanos , Período Intraoperatório , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Neoplasias Pancreáticas/cirurgia , Modelos de Riscos Proporcionais , Radioterapia Adjuvante , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...