Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Oncologist ; 25(5): 382-e776, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31857446

RESUMO

LESSONS LEARNED: Radioembolization with yttrium-90 resin microspheres can be combined safely with full doses of durvalumab and tremelimumab in patients with metastatic colorectal cancer. Regional radioembolization with yttrium-90 resin microspheres did not result in any hepatic or extrahepatic responses to a combination of durvalumab and tremelimumab. The lack of immunomodulatory responses to yttrium-90 on biopsies before and after treatment rules out a potential role for this strategy in converting a "cold tumor" into an "inflamed," immune responsive tumor. BACKGROUND: PD-1 inhibitors have been ineffective in microsatellite stable (MSS) metastatic colorectal cancer (CRC). Preclinical models suggest that radiation therapy may sensitize MSS CRC to PD-1 blockade. METHODS: Patients with MSS metastatic CRC with liver-predominant disease who progressed following at least one prior line of treatment were treated with yttrium-90 (Y90) radioembolization to the liver (SIR-Spheres; Sirtex, Woburn, MA) followed 2-3 weeks later by the combination of durvalumab and tremelimumab. A Simon two-stage design was implemented, with a planned expansion to 18 patients if at least one response was noted in the first nine patients. RESULTS: Nine patients enrolled in the first stage of the study, all with progressive disease (PD) during or after their first two cycles of treatment. Per preplanned design, the study was closed because of futility. No treatment-related grade 3 or greater toxicities were recorded. Correlative studies with tumor biopsies showed low levels of tumor-infiltrating lymphocyte (TIL) infiltration in tumor cancer islands before and after Y90 radioembolization. CONCLUSION: Y90 radioembolization can be added safely to durvalumab and tremelimumab but did not promote tumor-directed immune responses against liver-metastasized MSS CRC.


Assuntos
Neoplasias Colorretais , Embolização Terapêutica , Neoplasias Hepáticas , Anticorpos Monoclonais , Anticorpos Monoclonais Humanizados , Neoplasias Colorretais/tratamento farmacológico , Estudos de Viabilidade , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Repetições de Microssatélites , Resultado do Tratamento , Radioisótopos de Ítrio
2.
Blood ; 129(1): 114-125, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27760761

RESUMO

Attenuated poxvirus modified vaccinia Ankara (MVA) is a useful viral-based vaccine for clinical investigation, because of its excellent safety profile and property of inducing potent immune responses against recombinant (r) antigens. We developed Triplex by constructing an rMVA encoding 3 immunodominant cytomegalovirus (CMV) antigens, which stimulates a host antiviral response: UL83 (pp65), UL123 (IE1-exon4), and UL122 (IE2-exon5). We completed the first clinical evaluation of the Triplex vaccine in 24 healthy adults, with or without immunity to CMV and vaccinia virus (previous DryVax smallpox vaccination). Three escalating dose levels (DL) were administered IM in 8 subjects/DL, with an identical booster injection 28 days later and 1-year follow-up. Vaccinations at all DL were safe with no dose-limiting toxicities. No vaccine-related serious adverse events were documented. Local and systemic reactogenicity was transient and self-limiting. Robust, functional, and durable Triplex-driven expansions of CMV-specific T cells were detected by measuring T-cell surface levels of 4-1BB (CD137), binding to CMV-specific HLA multimers, and interferon-γ production. Marked and durable CMV-specific T-cell responses were also detected in Triplex-vaccinated CMV-seronegatives, and in DryVax-vaccinated subjects. Long-lived memory effector phenotype, associated with viral control during CMV primary infection, was predominantly found on the membrane of CMV-specific and functional T cells, whereas off-target vaccine responses activating memory T cells from the related herpesvirus Epstein-Barr virus remained undetectable. Combined safety and immunogenicity results of MVA in allogeneic hematopoietic stem cell transplant (HCT) recipients and Triplex in healthy adults motivated the initiation of a placebo-controlled multicenter trial of Triplex in HCT patients. This trial was registered at www.clinicaltrials.gov as #NCT02506933.


Assuntos
Antígenos Virais/imunologia , Vacinas contra Citomegalovirus/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Adulto , Citomegalovirus , Vacinas contra Citomegalovirus/efeitos adversos , Feminino , Humanos , Proteínas Imediatamente Precoces/imunologia , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/imunologia , Transativadores/imunologia , Vacinas de DNA , Proteínas da Matriz Viral/imunologia , Vacinas Virais , Adulto Jovem
3.
Cancer ; 123(23): 4566-4573, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28832978

RESUMO

BACKGROUND: Targeting the vascular endothelial growth factor (VEGF) pathway has improved outcomes in metastatic renal cell carcinoma (RCC); however, resistance inevitably occurs. CD105 (endoglin) is an angiogenic pathway that is strongly upregulated after VEGF inhibition, potentially contributing to resistance. The authors tested whether TRC105, a monoclonal antibody against endoglin, impacted disease control in patients with previously treated RCC who were receiving bevacizumab. METHODS: Eligible patients with metastatic RCC who had previously received 1 to 4 prior lines of therapy, including VEGF-targeted agents, were randomized 1:1 to receive bevacizumab 10 mg/kg intravenously every 2 weeks (arm A) or the same plus TRC105 10 mg/kg intravenously every 2 weeks (arm B). The primary endpoint was progression-free survival (PFS) at 12 and 24 weeks. Correlative studies included serum transforming growth factor ß (TGFß) and CD105 levels as well as tissue immunostaining for TGFß receptors. RESULTS: Fifty-nine patients were enrolled (28 on arm A and 31 on arm B), and 1 patient on each arm had a confirmed partial response. The median PFS for bevacizumab alone was 4.6 months compared with 2.8 for bevacizumab plus TRC105 (P = .09). Grade ≥ 3 toxicities occurred in 16 patients (57%) who received bevacizumab compared with 19 (61%) who received bevacizumab plus TRC105 (P = .9). Baseline serum TGFß levels below the median (<10.6 ng/mL) were associated with longer median PFS (5.6 vs 2.1 months; P = .014). CONCLUSIONS: TRC105 failed to improve PFS when added to bevacizumab. TGFß warrants further study as a biomarker in RCC. Cancer 2017;123:4566-4573. © 2017 American Cancer Society.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Papilar/tratamento farmacológico , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/administração & dosagem , Bevacizumab/administração & dosagem , Carcinoma Papilar/secundário , Carcinoma de Células Renais/secundário , Feminino , Seguimentos , Humanos , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Taxa de Sobrevida
4.
PLoS Pathog ; 10(11): e1004524, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25412505

RESUMO

Human Cytomegalovirus (HCMV) utilizes two different pathways for host cell entry. HCMV entry into fibroblasts requires glycoproteins gB and gH/gL, whereas HCMV entry into epithelial and endothelial cells (EC) requires an additional complex composed of gH, gL, UL128, UL130, and UL131A, referred to as the gH/gL-pentamer complex (gH/gL-PC). While there are no established correlates of protection against HCMV, antibodies are thought to be important in controlling infection. Neutralizing antibodies (NAb) that prevent gH/gL-PC mediated entry into EC are candidates to be assessed for in vivo protective function. However, these potent NAb are predominantly directed against conformational epitopes derived from the assembled gH/gL-PC. To address these concerns, we constructed Modified Vaccinia Ankara (MVA) viruses co-expressing all five gH/gL-PC subunits (MVA-gH/gL-PC), subsets of gH/gL-PC subunits (gH/gL or UL128/UL130/UL131A), or the gB subunit from HCMV strain TB40/E. We provide evidence for cell surface expression and assembly of complexes expressing full-length gH or gB, or their secretion when the corresponding transmembrane domains are deleted. Mice or rhesus macaques (RM) were vaccinated three times with MVA recombinants and serum NAb titers that prevented 50% infection of human EC or fibroblasts by HCMV TB40/E were determined. NAb responses induced by MVA-gH/gL-PC blocked HCMV infection of EC with potencies that were two orders of magnitude greater than those induced by MVA expressing gH/gL, UL128-UL131A, or gB. In addition, MVA-gH/gL-PC induced NAb responses that were durable and efficacious to prevent HCMV infection of Hofbauer macrophages, a fetal-derived cell localized within the placenta. NAb were also detectable in saliva of vaccinated RM and reached serum peak levels comparable to NAb titers found in HCMV hyperimmune globulins. This vaccine based on a translational poxvirus platform co-delivers all five HCMV gH/gL-PC subunits to achieve robust humoral responses that neutralize HCMV infection of EC, placental macrophages and fibroblasts, properties of potential value in a prophylactic vaccine.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecções por Citomegalovirus , Vacinas contra Citomegalovirus , Citomegalovirus , Complexos Multiproteicos , Proteínas do Envelope Viral , Animais , Citomegalovirus/genética , Citomegalovirus/imunologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/genética , Vacinas contra Citomegalovirus/imunologia , Feminino , Humanos , Macaca mulatta , Camundongos , Camundongos Endogâmicos BALB C , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
5.
J Virol ; 87(3): 1322-32, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23152525

RESUMO

Neutralizing antibodies (NAb) are important for interfering with horizontal transmission of human cytomegalovirus (HCMV) leading to primary and congenital HCMV infection. Recent findings have shown that a pentameric virion complex formed by the glycoproteins gH/gL, UL128, UL130, and UL131A (UL128C) is required for HCMV entry into epithelial/endothelial cells (Epi/EC) and is the target of potent NAb in HCMV-seropositive individuals. Using bacterial artificial chromosome technology, we have generated a modified vaccinia Ankara virus (MVA) that stably coexpresses all 5 rhesus CMV (RhCMV) proteins homologous to HCMV UL128C, termed MVA-RhUL128C. Coimmunoprecipitation confirmed the interaction of RhgH with the other 4 RhCMV subunits of the pentameric complex. All 8 RhCMV-naïve rhesus macaques (RM) vaccinated with MVA-RhUL128C developed NAb that blocked infection of monkey kidney epithelial cells (MKE) and rhesus fibroblasts. NAb titers induced by MVA-RhUL128C measured on both cell types at 2 to 6 weeks postvaccination were comparable to levels observed in naturally infected RM. In contrast, MVA expressing a subset of RhUL128C proteins or RhgB glycoprotein only minimally stimulated NAb that inhibited infection of MKE. In addition, following subcutaneous RhCMV challenge at 8 weeks postvaccination, animals vaccinated with MVA-RhUL128C showed reduced plasma viral loads. These results indicate that MVA expressing the RhUL128C induces NAb inhibiting RhCMV entry into both Epi/EC and fibroblasts and limits RhCMV replication in RM. This novel approach is the first step in developing a prophylactic HCMV vaccine designed to interfere with virus entry into major cell types permissive for viral replication, a required property of an effective vaccine.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra Citomegalovirus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/administração & dosagem , DNA Viral/química , DNA Viral/genética , Portadores de Fármacos , Células Epiteliais/virologia , Fibroblastos/virologia , Vetores Genéticos , Macaca mulatta , Dados de Sequência Molecular , Análise de Sequência de DNA , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Proteínas do Envelope Viral/administração & dosagem , Viremia/prevenção & controle , Internalização do Vírus
6.
Blood ; 118(8): 2159-69, 2011 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-21719601

RESUMO

The serine proteases, neutrophil elastase (HNE) and proteinase 3 (PR3), are aberrantly expressed in human myeloid leukemias. T-cell responses to these proteins have been correlated with remission in patients with chronic myeloid leukemia (CML). Human PR3/HNE-specific CD8(+) T cells predominantly recognize a nonameric HLA-A2-restricted T-cell epitope called PR1 which is conserved in both Ags. However, CML patients have CD8(+) T cells in peripheral blood recognizing an additional HLA-A2 epitope termed PR2. To assess immunologic properties of these Ags, novel recombinant vaccinia viruses (rVV) expressing PR3 and HNE were evaluated in HLA-A2 transgenic (Tg) mice (HHDII). Immunization of HHDII mice with rVV-PR3 elicited a robust PR3-specific CD8(+) T-cell response dominated by recognition of PR2, with minimal recognition of the PR1 epitope. This result was unexpected, because the PR2 peptide has been reported to bind poorly to HLA. To account for these findings, we proposed that HHDII mice negatively selected PR1-specific T cells because of the presence of this epitope within murine PR3 and HNE, leading to immunodominance of PR2-specific responses. PR2-specific splenocytes are cytotoxic to targets expressing naturally processed PR3, though PR1-specific splenocytes are not. We conclude that PR2 represents a functional T-cell epitope recognized in mice and human leukemia patients. These studies are registered at www.clinicaltrials.gov as NCT00716911.


Assuntos
Epitopos de Linfócito T/imunologia , Antígeno HLA-A2/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Grânulos Citoplasmáticos/enzimologia , Grânulos Citoplasmáticos/imunologia , Epitopos de Linfócito T/genética , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Transplante de Células-Tronco Hematopoéticas , Humanos , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Elastase de Leucócito/genética , Elastase de Leucócito/imunologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mieloblastina/genética , Mieloblastina/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Homologia de Sequência de Aminoácidos , Linfócitos T Citotóxicos/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia
7.
J Infect Dis ; 205(8): 1294-304, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22402037

RESUMO

BACKGROUND: It has been reported that cytomegalovirus (CMV) pp65-specific T cells can protect hematopoietic cell transplant (HCT) recipients from CMV complications. Two candidate CMV peptide vaccines composed of the HLA A*0201 pp65(495-503) cytotoxic CD8(+) T-cell epitope fused to 2 different universal T-helper epitopes (either the synthetic Pan DR epitope [PADRE] or a natural Tetanus sequence) were clinically evaluated for safety and ability to elicit pp65 T cells in HLA A*0201 healthy volunteers. METHODS: Escalating doses (0.5, 2.5, 10 mg) of PADRE or Tetanus pp65(495-503) vaccines with (30 adults) or without (28 adults) PF03512676 adjuvant were administered by subcutaneous injection every 3 weeks for a total of 4 injections. RESULTS: No serious adverse events were reported, although vaccines used in combination with PF03512676 had enhanced reactogenicity. Ex vivo responses were detected by flow cytometry exclusively in volunteers who received the vaccine coadministered with PF03512676. In addition, using a sensitive in vitro stimulation system, vaccine-elicited pp65(495-503) T cells were expanded in 30% of volunteers injected solely with the CMV peptides and in all tested subjects receiving the vaccines coinjected with PF03512676. CONCLUSIONS: Acceptable safety profiles and vaccine-driven expansion of pp65(495-503) T cells in healthy adults support further evaluation of CMV peptide vaccines combined with PF03512676 in the HCT setting. CLINICAL TRIALS REGISTRATION: NCT00722839.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Infecções por Citomegalovirus/prevenção & controle , Vacinas contra Citomegalovirus/imunologia , Vacinas Antimaláricas/imunologia , Oligodesoxirribonucleotídeos/administração & dosagem , Toxoide Tetânico/imunologia , Adjuvantes Imunológicos/efeitos adversos , Adolescente , Adulto , Sequência de Aminoácidos , Linfócitos T CD8-Positivos/fisiologia , Vacinas contra Citomegalovirus/administração & dosagem , Vacinas contra Citomegalovirus/efeitos adversos , Relação Dose-Resposta Imunológica , Epitopos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Oligodesoxirribonucleotídeos/efeitos adversos , Proteínas Recombinantes/imunologia , Toxoide Tetânico/administração & dosagem , Toxoide Tetânico/efeitos adversos , Vacinas Sintéticas , Adulto Jovem
8.
Blood ; 113(25): 6465-76, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19369230

RESUMO

Reconstitution of cytomegalovirus (CMV)-specific CD8(+) T cells is essential to the control of CMV infection in CMV-positive recipients (R(+)) after allogeneic hematopoietic stem cell transplantation (HCT). Six-color flow cytometry was used to assess the functional profile of CMV-specific CD8(+) T cells in 62 of 178 R(+) HCT recipients followed virologically for CMV reactivation. R(+) recipients receiving grafts from CMV-negative donors (D(-); D(-)/R(+)) reconstituted fewer multifunctional CD8(+) T cells expressing tumor necrosis factor-alpha (TNF-alpha), macrophage inflammatory protein-1beta (MIP-1beta), and CD107 in addition to interferon-gamma (IFN-gamma), compared with D(+)/R(+) recipients. Unlike monofunctional CD8(+) T cells secreting IFN-gamma, which were abundantly generated during CMV reactivation in D(-)/R(+) recipients, the relative lack of multifunctional CD8(+) T cells persisted until at least 1 year post-HCT. D(-)/R(+) recipients were more likely to require recurrent and prolonged use of antivirals. These findings were robust to statistical adjustment for pretransplant factors, as well as for posttransplant factors including graft-versus-host disease (GVHD) and its treatment by steroids. These analyses suggest that D(+)/R(+) transplants, on average, generate higher levels of multifunctional CMV-specific T cells and require less antiviral therapy compared with D(-)/R(+) HCT recipients. These results highlight the benefit of D(+) donors in improving outcomes of R(+) HCT recipients by reducing the duration and recurrent need of antiviral treatment, aided by increased levels of multifunctional CMV-specific T cells.


Assuntos
Transplante de Medula Óssea/estatística & dados numéricos , Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/isolamento & purificação , Transplante de Células-Tronco de Sangue Periférico/estatística & dados numéricos , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/imunologia , Doadores de Tecidos , Transplante Homólogo/estatística & dados numéricos , Adulto , Antivirais/efeitos adversos , Antivirais/uso terapêutico , Transplante de Medula Óssea/efeitos adversos , Linfócitos T CD8-Positivos/metabolismo , Estudos de Coortes , Citocinas/metabolismo , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/fisiopatologia , Infecções por Citomegalovirus/transmissão , Vacinas contra Citomegalovirus , Transmissão de Doença Infecciosa , Feminino , Sobrevivência de Enxerto/imunologia , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/cirurgia , Humanos , Imunidade Celular , Masculino , Pessoa de Meia-Idade , Transplante de Células-Tronco de Sangue Periférico/efeitos adversos , Fosfoproteínas/imunologia , Irmãos , Transplante Homólogo/efeitos adversos , Resultado do Tratamento , Proteínas da Matriz Viral/imunologia , Ativação Viral
9.
Transpl Int ; 24(9): 920-31, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21672050

RESUMO

Antiviral prophylaxis has proved successful for prevention of cytomegalovirus (CMV) disease in solid organ transplant (SOT) patients; though emerging data suggest that antiviral agents interfere with immunity, and may inhibit immune priming. In this context, we investigated levels and phenotype of primary CMV-specific immune responses that developed during antiviral prophylaxis in a cohort of CMV seronegative recipients (R(-) ) of a SOT from a seropositive donor (D(+) ). We longitudinally monitored CMV viral load, antibodies and levels of the negative immuno-modulator IL-10. PBMC were stimulated with CMV-specific peptide libraries to measure CD137 activation marker on CMV-specific T-cells and levels of PD-1 receptor, which is over expressed on exhausted T-cells. Unexpectedly, the majority (13/18) of D(+) R(-) patients who developed a primary CMV response showed early post-transplant CMV-specific responses, though levels of PD-1 on CMV-specific T-cells remained elevated throughout prophylaxis. A strong inverse association was found between levels of plasma IL-10 and CMV-specific cellular immune responses. Our study suggests that during prophylaxis, subclinical CMV infection might have occurred in the D(+) R(-) patients, and primary CMV-specific responses were detected early post-transplant when levels of plasma IL-10 were low. Extended prophylaxis or antiviral treatment did not appear to suppress CMV-specific antibodies or T-cells, which, however, showed exhaustion phenotypes.


Assuntos
Antivirais/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/imunologia , Ganciclovir/análogos & derivados , Transplante de Rim/métodos , Transplante de Fígado/métodos , Adulto , Idoso , Antígenos Virais/imunologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/prevenção & controle , Feminino , Ganciclovir/uso terapêutico , Humanos , Proteínas Imediatamente Precoces/imunologia , Interleucina-10/biossíntese , Interleucina-10/imunologia , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/imunologia , Linfócitos T/imunologia , Linfócitos T/virologia , Valganciclovir , Carga Viral , Proteínas da Matriz Viral/imunologia
10.
Int J Clin Oncol ; 16(5): 494-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21431345

RESUMO

BACKGROUND: This study was conducted to evaluate the efficacy of the epidermal growth factor receptor tyrosine kinase inhibitor gefitinib with pegylated-IFNα (PEG-IFNα) in patients with advanced renal cell carcinoma. METHODS: Progression-free survival (PFS) rate at 6 months >50% was considered promising for further evaluation. Patients with unresectable or metastatic disease, unlimited prior therapies, and adequate performance status and end-organ function were eligible. PEG-IFNα was dosed subcutaneously once weekly (initially 6 µg/kg/week, later reduced to 4 µg/kg/week) for 12 weeks. Gefitinib was given 250 mg orally once daily until progression or intolerance. RESULTS: Twenty-one patients were accrued. Fourteen patients had a prior nephrectomy, and twelve had prior systemic therapy. The 6-month PFS was 29% (95%CI 15-56%). Best responses by RECIST criteria: complete, partial (1, plus 3 unconfirmed) stable (Uhlman et al. Clin Cancer Res 1:913-920, 1995), and progression (Sirotnak et al. Clin Cancer Res 6:4885-4892, 2000). Response duration: complete response (35+ months) and partial response (2, 3, 3, 37 months). Median PFS and overall survival were 5.3 (95%CI 3-10.1) and 13.6 (95%CI 10.3-NA) months, respectively. Most common toxicities included myelosuppression, rash, and nausea. CONCLUSIONS: Although generally well tolerated, gefitinib plus PEG-IFNα did not meet the pre-specified 6-month PFS rate >50%. Further evaluation of similar regimens would require appropriate molecular selection of subjects most likely to benefit. Thus, preclinical studies to determine candidate predictive markers for this combination are warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Carcinoma de Células Renais/tratamento farmacológico , Interferon-alfa/administração & dosagem , Neoplasias Renais/tratamento farmacológico , Polietilenoglicóis/administração & dosagem , Quinazolinas/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carcinoma de Células Renais/patologia , Intervalo Livre de Doença , Feminino , Gefitinibe , Humanos , Interferon-alfa/efeitos adversos , Neoplasias Renais/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Polietilenoglicóis/efeitos adversos , Quinazolinas/efeitos adversos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos
11.
Curr Probl Cancer ; 44(6): 100583, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32446637

RESUMO

BACKGROUND: There is increasing use of Phase I statistical designs to find a dose that causes rapidly emerging and particularly concerning severe or life-threatening toxicities (dose-limiting toxicities, DLTs) in a specified percent of patients most commonly 25%. While a convenient statistical framework, the foundation for selecting any specified target DLT rate, and its relevance to the recommended Phase II dose is generally lacking. METHOD: We surveyed 78 medical oncologists, most (69%) with experience as a principal investigator on a Phase I study, to ascertain their opinions related to this approach to Phase I studies and the targets often chosen. RESULTS: Eighty-seven percent of respondents preferred severe toxicities in only 5%-10% of patients, consistent with 58% of respondents noting that 10% or fewer patients experience severe toxicities in the first cycle with standard outpatient treatments. The survey also documented in an example that the majority (62%) of physicians modify their patient selection during the conduct of the study based on observed toxicity and 78% note that higher toxicity is acceptable in patients where a cure is more likely. CONCLUSION: DLT-target rate designs search for a single target that is rarely well-supported in a patient population that is not stable. The most common target used is inconsistent with the toxicity of most clinically used drugs and investigator preference and can lead to the pursuit of unacceptable doses. Use of Phase I trial designs with a target DLT rate should be limited to settings with a well-justified target and should specify how the target relates to the recommended Phase II dose.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Ensaios Clínicos Fase I como Assunto/normas , Cálculos da Dosagem de Medicamento , Neoplasias/tratamento farmacológico , Oncologistas/estatística & dados numéricos , Projetos de Pesquisa/normas , Relação Dose-Resposta a Droga , Humanos , Neoplasias/patologia , Seleção de Pacientes , Inquéritos e Questionários
12.
Anticancer Drugs ; 20(3): 179-84, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19396016

RESUMO

Prostate cancer cells undergo neuroendocrine differentiation during androgen deprivation and secrete neuropeptides, hence activating androgen receptor-regulated genes. Src-family protein kinases are involved in neuropeptide-induced prostate cancer growth and migration. A phase II trial of AZD0530, an oral Src-family kinase inhibitor, in patients with advanced castration resistant prostate cancer was conducted. The primary endpoint was prostate cancer-specific antigen (PSA) response rate, defined as a 30% or greater decrease. A two-stage Simon design was used. Eligibility criteria included documentation of castration resistance (including antiandrogen withdrawal), adequate end-organ function, and performance status, and not more than one prior taxane-based chemotherapy regimen. AZD0530 was given at 175 mg orally once daily continuously. Rapid accrual led to 28 patients registering in the first stage. Median age was 67 years. Sixteen patients had performance status (PS) 0, eight patients had PS 1, and four patients had PS 2. Nine patients (32%) had prior docetaxel-based chemotherapy. Five patients had transient PSA reductions not meeting PSA response criteria. Median progression-free survival time was 8 weeks. Treatment was generally well tolerated. AZD0530, a potent oral Src kinase inhibitor, is feasible and tolerable in this pretreated patient population but possessed little clinical efficacy as monotherapy. Strong preclinical evidence warrants further investigation of AZD0530 in earlier-stage prostate cancer or as combination therapy.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Benzodioxóis/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Quinazolinas/uso terapêutico , Quinases da Família src/antagonistas & inibidores , Adenocarcinoma/sangue , Adenocarcinoma/cirurgia , Idoso , Idoso de 80 Anos ou mais , Antagonistas de Androgênios/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Benzodioxóis/efeitos adversos , Benzodioxóis/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Terapia Combinada , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos , Gastroenteropatias/induzido quimicamente , Humanos , Linfopenia/induzido quimicamente , Masculino , Pessoa de Meia-Idade , Orquiectomia , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/cirurgia , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/efeitos adversos , Quinazolinas/farmacologia
13.
Clin Genitourin Cancer ; 5(7): 433-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18272025

RESUMO

BACKGROUND: Perifosine is an oral alkylphospholipid that inhibits cancer cell growth through decreased Akt phosphorylation. We conducted a phase II trial of perifosine in patients with biochemically recurrent, hormone-sensitive prostate cancer. PATIENTS AND METHODS: Eligible patients had histologically confirmed prostate cancer, previous prostatectomy and/or radiation therapy, and rising prostate-specific antigen (PSA) without radiographic evidence of metastasis. Previous androgen deprivation therapy < 9 months in duration (completed >or= 1 year before registration) was allowed. The primary endpoint was PSA response, defined as a decrease by >or= 50% from the pretreatment value. Treatment was composed of a loading dose of perifosine 900 mg orally on day 1, then 100 mg daily starting 24 hours later. RESULTS: Of 25 patients, 24 were evaluable for response. After a median follow-up of 8 months, 5 patients (20%) had a reduction in serum PSA levels, but none met criteria for PSA response. Three patients immediately progressed with no response to therapy. Median progression-free survival was 6.64 months (range, 4.53-12.81 months). No change in the PSA doubling time (7 months) was observed before and after treatment initiation. Dose-limiting toxicities (all grade 3) included hyponatremia, arthritis, hyperuricemia, and photophobia. CONCLUSION: Although well tolerated, perifosine did not meet prespecified PSA criteria for response as a single agent in biochemically recurrent prostate cancer. However, 20% of patients had evidence of PSA reduction, suggesting modest single-agent clinical activity. The role of perifosine in combination with androgen deprivation or chemotherapy is currently under investigation.


Assuntos
Recidiva Local de Neoplasia/tratamento farmacológico , Fosforilcolina/análogos & derivados , Neoplasias da Próstata/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Idoso , Antagonistas de Androgênios/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilcolina/efeitos adversos , Fosforilcolina/uso terapêutico
14.
Clin Cancer Res ; 12(5): 1556-63, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16533781

RESUMO

BACKGROUND: BMS-275291 is a selective matrix metalloproteinase inhibitor (MMPI) that does not inhibit sheddases implicated in the dose-limiting arthritis of older MMPIs. We conducted a randomized phase II trial of two doses of BMS-275291 (1,200 versus 2,400 mg) in hormone-refractory prostate cancer (HRPC) patients with bone metastases to probe for a dose-response relationship and to assess differential toxicities. Serial serum and urine specimens were collected to assess for markers of bone metabolism. METHODS: The primary end point was 4-month progression-free survival (PFS). Eligibility criteria included documentation of androgen-independent disease (including anti-androgen withdrawal), skeletal metastasis, adequate end-organ function and performance status, and no more than one prior chemotherapy regimen. Patients were randomized to 1,200 mg orally once daily (arm A) or 1,200 mg orally twice daily (arm B). Response was assessed every 56 days. RESULTS: Eighty patients were enrolled: 39 in arm A and 41 in arm B. There were no responders by prostate-specific antigen or measurable disease to treatment. Stable disease was noted at 8 weeks in 39% of patients in arm A and in 17% of patients in arm B. Progression of disease at 8 weeks was seen in 61% of patients in arm A versus 83% of patients in arm B. Median survival time was 21.6 months (95% confidence interval, 17.5; not reached), whereas median PFS time was 1.8 months (95% confidence interval 1.74; 2) for all patients. Patients in arm A had a median survival time that was not reached, whereas patients on arm B has a median survival time of 21 months (P = 0.2). PFS at 4 months favored arm A: 22% versus 10% (log-rank, P = 0.008). Grade 3 toxicities occurred in 5 (13%) patients in arm A and in 9 (22%) patients in arm B. Grade 4 toxicities were uncommon (only 4% of patients): one each of thrombosis, fatigue, and motor neuropathy was seen in the arm B. Bone marker studies showed that baseline serum levels of N-telopeptide, osteocalcin, procollagen I NH2-terminal propeptide, and PICP had prognostic significance for PFS and/or overall survival. CONCLUSIONS: Regardless of dose schedule, BMS-275291 was well tolerated in HRPC patients and had no dose-limiting arthritis. Toxicities differed modestly according to the dose schedule employed. As overall survival and PFS favored the once daily schedule, this dose schedule is recommended for future studies. Baseline markers of bone metabolism may have prognostic value in HRPC patients with bone metastases.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Imidazóis/uso terapêutico , Inibidores de Metaloproteinases de Matriz , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Neoplasias Ósseas/secundário , Intervalo Livre de Doença , Esquema de Medicação , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Hormônio-Dependentes/patologia , Neoplasias da Próstata/patologia , Taxa de Sobrevida , Resultado do Tratamento
15.
Exp Hematol ; 34(4): 497-507, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16569596

RESUMO

OBJECTIVE: Adoptive cellular therapy of cytomegalovirus (CMV)-specific T cells in allogeneic hematopoietic stem cell transplantation (HSCT) patients is a promising approach for controlling CMV viremia and its morbidity. We sought to develop a clinically suitable strategy to dually expand infusible CD8(+) and CD4(+) T-cell subsets specific for CMV. METHODS: Polyclonal CMV T-cell lines were generated using peripheral blood mononuclear cell (PBMCs) treated with synthetic single-stranded CpG motif-containing oligodeoxynucleotides (ODNs) and infected with recombinant (r) modified vaccinia Ankara (MVA) expressing CMV antigens. Cultures derived from 12 healthy CMV-positive donors were analyzed using chromium release and lymphoproliferation assays, intracellular staining for interferon-gamma (IFN-gamma), and HLA tetramers. RESULTS: A 3-day incubation with a combination of ODN 2006 and 2216 was found to reproducibly generate a highly rMVA infectable population of PBMCs with concomitant high expression of CMV antigens. CpG ODN-treated autologous PBMCs infected with rMVA elicited a 30-fold average expansion of both CMV-specific CD4(+) and CD8(+) T cells in 10 days. The enriched T-cell populations showed minimal alloreactivity, high levels of CMV-specific HLA class I tetramer binding, cytotoxic activity, and IFN-gamma production from both CD8(+) and CD4(+) T cells. CONCLUSIONS: The ability to quickly produce autologous professional antigen-presenting cells, capable of stimulating clinically useful amounts of CMV-specific CD4(+) and CD8(+) T-cell lines, enhances the attractiveness of using rMVA for immunotherapeutic interventions to manage HSCT-related CMV disease.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Ativação Linfocitária/imunologia , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/virologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/transplante , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ilhas de CpG/imunologia , Citomegalovirus/genética , Infecções por Citomegalovirus/etiologia , Infecções por Citomegalovirus/terapia , Antígenos HLA/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Imunoterapia Adotiva/métodos , Interferon gama/imunologia , Ativação Linfocitária/efeitos dos fármacos , Transfusão de Linfócitos/métodos , Oligodesoxirribonucleotídeos/farmacologia , Transplante Homólogo , Vaccinia virus/genética , Vaccinia virus/imunologia
16.
J Clin Oncol ; 23(22): 4876-80, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16009960

RESUMO

PURPOSE: To determine whether floxuridine (FUDR) can be delivered with low hepatic toxicity through the portal vein (PV) as an adjuvant to surgically treated colorectal metastases. PATIENTS AND METHODS: Fifty-one patients undergoing complete resection and/or ablation for colorectal hepatic metastases were prospectively enrolled at a National Cancer Institute-designated comprehensive cancer center. Two sequential phase II trials were performed. Each trial included complete surgical treatment followed by sequential, alternating (22 patients) or concurrent (29 patients) regional PV FUDR and systemic fluorouracil (FU) with leucovorin chemotherapy. RESULTS: Fifty percent of patients were male. The mean age at diagnosis was 57 years. The mean number of lesions resected was three (range, one to 11 lesions). The stage at diagnosis was II, III, and IV in 16.9%, 52.8%, and 28.3% of patients, respectively. One- and 3-year overall survival rates were 92.7% and 41.8%, respectively. The 1- and 3-year disease-free survival rates were 64.5% and 19%, respectively. The site of first recurrence was hepatic in 35.9% of patients. Treatment was terminated early in 24 patients (17 patients progressed, two refused treatment, and five had nonhepatic toxicities). Fifty-five percent of patients received 75% to 100% of the planned FUDR courses, and 72% received greater than 50% of the planned FUDR dose. Only four patients required dose reductions of FUDR because of grade 3 hepatic toxicity. No patient required biliary stenting or had discontinuation of PV infusion because of hepatic toxicity. CONCLUSION: The delivery of PV FUDR and FU with leucovorin can be performed with a high percentage of expected drug delivery and a low drug-induced hepatic toxicity rate, while achieving acceptable overall and disease-free survival.


Assuntos
Antimetabólitos Antineoplásicos/efeitos adversos , Antimetabólitos Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Floxuridina/efeitos adversos , Floxuridina/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Adulto , Idoso , Antimetabólitos Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Quimioterapia Adjuvante , Intervalo Livre de Doença , Feminino , Floxuridina/administração & dosagem , Fluoruracila/administração & dosagem , Humanos , Infusões Intravenosas , Leucovorina/administração & dosagem , Neoplasias Hepáticas/cirurgia , Masculino , Pessoa de Meia-Idade , Veia Porta
17.
Clin Cancer Res ; 11(12): 4444-50, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15958629

RESUMO

BACKGROUND: UCN-01 (7-hydroxy-staurosporine) is a novel antineoplastic agent targeting cyclin-dependent kinases, which shows potent in vitro and in vivo activity against a broad range of tumor types. Our group has previously shown that UCN-01 potentiates the apoptotic response of agents such as cisplatin in vitro by preventing sequence-specific abrogation of G2 arrest caused by DNA-damaging chemotherapies. PATIENTS AND METHODS: This National Cancer Institute-sponsored phase I trial was designed to determine the safety, maximum tolerated dose, and pharmacokinetics of escalating doses of cisplatin in combination with UCN-01 in patients with advanced malignant solid tumors, as well as to do molecular correlative studies on tumor specimens. Cisplatin was infused over 1 hour before UCN-01 (45 mg/m2/d) given as a 72-hour continuous infusion. Escalation of cisplatin was planned through five dose levels at 20, 30, 45, 60, and 75 mg/m2. RESULTS: Ten patients were accrued. Accrual was halted at dose level 2 (cisplatin, 30 mg/m2) due to dose-limiting toxicities consisting of grade 5 sepsis with respiratory failure associated with grade 3 creatinine (one patient) and grade 3 atrial fibrillation (one patient). Plasma and salivary pharmacokinetics of UCN-01 were unaffected by cisplatin. Pretreatment and posttreatment tumor biopsies showed that UCN-01 was active against a key molecular target, the checkpoint kinase Chk1. CONCLUSIONS: This phase I trial failed to achieve targeted therapeutic dose levels of cisplatin when combined with prolonged infusion UCN-01. However, because preclinical data indicate that UCN-01 potentiates response to platinum, further studies with alternative dose schedules of the combination, or with other platinum analogues, are warranted.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias/tratamento farmacológico , Estaurosporina/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Área Sob a Curva , Fibrilação Atrial/induzido quimicamente , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Cisplatino/farmacocinética , Ciclina B/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Transtornos de Deglutição/induzido quimicamente , Relação Dose-Resposta a Droga , Feminino , Humanos , Hiponatremia/induzido quimicamente , Infecções/induzido quimicamente , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Proteínas Quinases/metabolismo , Insuficiência Renal/induzido quimicamente , Estaurosporina/administração & dosagem , Estaurosporina/efeitos adversos , Estaurosporina/farmacocinética , Taquicardia/induzido quimicamente , Resultado do Tratamento , Proteínas Supressoras de Tumor/metabolismo , Fosfatases cdc25/metabolismo
18.
Cancer Res ; 64(15): 5407-14, 2004 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15289349

RESUMO

The p53 gene product is overexpressed by almost 50% of cancers, making it an ideal target for cancer immunotherapy. We previously demonstrated rejection of established p53-overexpressing tumors without stimulating autoimmunity by immunization with modified vaccinia Ankara-expressing murine p53 (MVAp53). Tumor rejection was enhanced through antibody-mediated CTL-associated antigen 4 (CTLA-4) blockade. We examined the role of synthetic oligodeoxynucleotides (ODN) containing unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG ODN) in enhancing MVAp53-mediated tumor rejection. CpG ODN with MVAp53 resulted in tumor rejection in BALB/c mice bearing poorly immunogenic 11A-1 murine mammary carcinomas or Meth A sarcomas and C57Bl/6 mice bearing MC-38 colon carcinomas. The effect was similar to that seen in tumor-bearing mice treated with MVAp53 along with CTLA-4 blockade. Monoclonal antibody depletion experiments demonstrated that the adjuvant effects of CpG ODN and CTLA-4 blockades were CD8 dependent. CpG ODN were partially natural killer cell dependent and ineffective in Toll-like Receptor 9(-/-) and interleukin 6(-/-) mice, whereas CTLA-4 blockade was partially CD4 dependent and functional in Toll-like Receptor 9(-/-) and interleukin 6(-/-) mice. In addition, when administered with MVAp53, both adjuvants enhanced p53-specific cytotoxicity and demonstrated an additive effect when combined. The combination of CpG ODN and CTLA-4 blockade worked synergistically to reject palpable 11A-1 and MC-38 tumors. These experiments demonstrate the potential for augmenting MVAp53-mediated antitumor immunity using CpG ODN and CTLA-4 blockade. This cell-free immunotherapy approach is a candidate for evaluation in cancer patients.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/terapia , Neoplasias Mamárias Animais/terapia , Sarcoma Experimental/terapia , Transdução de Sinais , Proteína Supressora de Tumor p53/farmacologia , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Animais , Anticorpos Monoclonais/metabolismo , Antígenos CD , Antígenos de Diferenciação/química , Antígeno CTLA-4 , Células Cultivadas , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Terapia Combinada , Cricetinae , Testes Imunológicos de Citotoxicidade , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Sinergismo Farmacológico , Feminino , Homozigoto , Humanos , Imunização , Interleucina-6/genética , Interleucina-6/fisiologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodesoxirribonucleotídeos/farmacologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Sarcoma Experimental/genética , Sarcoma Experimental/imunologia , Receptor Toll-Like 9 , Proteína Supressora de Tumor p53/genética
19.
Transplantation ; 100(1): 210-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26680375

RESUMO

BACKGROUND: Immune measurements that distinguish solid organ transplantation (SOT) recipients who control cytomegalovirus (CMV) infection from those who progress to CMV-disease (CMV-dz) may be clinically useful in guiding tailored prevention strategies. We previously reported that elevated plasma levels of the immune-modulator IL-10 are associated with late CMV-dz. Here we evaluate whether IL-10 levels measured soon after prophylaxis discontinuation are predictive of CMV-dz risk. METHODS: Plasma IL-10 levels were quantitatively measured by ELISA kit in 40 D/R SOT patients. All 40 D/R high-risk patients were prospectively followed for at least 12 months post-SOT: 13 subjects developed CMV-dz, all within 6 months of prophylaxis discontinuation. RESULTS: IL-10 was detectable at the first post-prophylaxis measurement for 11 of 13 subjects who developed CMV-dz. In contrast, IL-10 was detectable in only 6 of 27 CMV asymptomatic patients. Monitoring IL-10 plasma levels within 1 month prophylaxis suspension appeared to have clinically useful level of 85% sensitivity and 78% specificity. CONCLUSIONS: The exact role of IL-10 with its multiple immunoregulatory effects during CMV infection is not clear. Moreover, IL-10 production can be influenced by pathological and infectious contexts, and/or anti-rejection immunosuppressant therapy. Despite mechanisms of IL-10 dysregulation may substantially differ among SOT patients, our findings suggest that measurable plasma IL-10 soon after prophylaxis discontinuation may be an adequate indicator of subsequent CMV-dz. If a similar prognostic performance is confirmed in a larger D/R cohort, IL-10 plasma levels could be used to guide the length of prophylaxis, providing a clinically useful means to reduce the incidence of CMV-dz in high risk patients.


Assuntos
Antivirais/administração & dosagem , Infecções por Citomegalovirus/prevenção & controle , Monitoramento de Medicamentos/métodos , Ganciclovir/análogos & derivados , Interleucina-10/sangue , Transplante de Rim/efeitos adversos , Transplante de Fígado/efeitos adversos , Transplantados , Biomarcadores/sangue , Infecções por Citomegalovirus/sangue , Infecções por Citomegalovirus/diagnóstico , Infecções por Citomegalovirus/imunologia , Esquema de Medicação , Ensaio de Imunoadsorção Enzimática , Ganciclovir/administração & dosagem , Humanos , Valor Preditivo dos Testes , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento , Regulação para Cima , Valganciclovir
20.
Clin Cancer Res ; 10(2): 461-7, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14760066

RESUMO

PURPOSE: To determine if there is a beneficial effect of amifostine in preventing or reducing the neuropathy induced by high-dose paclitaxel. METHODS: Breast cancer patients receiving high-dose infusional paclitaxel (725 mg/m(2)/24 h) in combination with doxorubicin (165 mg/m(2)/96 h) and cyclophosphamide (100 mg/kg/2 h; ACT) were studied on two autologous peripheral blood stem cell transplant protocols, one with and one without amifostine (740 mg/m(2) administered over 10 min before and 12 h after initiation of the paclitaxel infusion). Patients were evaluated before ACT and 20-40 days later with neurological examination, a composite peripheral neuropathy score, peroneal and sural nerve conduction studies, and quantitative sensory testing. RESULTS: There was no significant difference in paclitaxel maximum concentration, systemic clearance, or area under the curve determinations. Narcotic requirement as well as recovery of hematopoietic counts were also similar in subjects with or without amifostine. After ACT was administered, there was a decrease in peroneal nerve compound muscle action potential amplitude and sural nerve sensory action potential amplitude, as well as an increase in vibratory and cold detection thresholds. Clinical composite peripheral neuropathy scores were similar despite amifostine treatment; and logarithm to the base 2 ratios post/pre ACT showed no significant effect of amifostine on peroneal nerve compound muscle action potential, sural nerve sensory action potential, vibratory detection thresholds, or cold detection thresholds. All subjects had acroparesthesias and lost their ankle deep-tendon reflexes after administration of ACT. CONCLUSIONS: Single high-dose paclitaxel produces predictable clinical and neurophysiological changes so that patients receiving high-dose therapy are ideal subjects to test the effectiveness of neuroprotective agents. Amifostine was ineffective in preventing or reducing the neurotoxicity of high-dose paclitaxel.


Assuntos
Amifostina/farmacologia , Paclitaxel/efeitos adversos , Paclitaxel/farmacologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/prevenção & controle , Adulto , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Área Sob a Curva , Neoplasias da Mama/tratamento farmacológico , Ciclofosfamida/farmacologia , Doxorrubicina/farmacologia , Sinergismo Farmacológico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Pessoa de Meia-Idade , Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Protetores contra Radiação/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa