Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Am Surg ; : 31348241241651, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38549202

RESUMEN

Retroperitoneal abscess as a sequela of penetrating trauma can pose a difficult clinical scenario for surgeons and literature to inform decision making is sparse. It is logical to follow a "step-up" approach applied to other etiologies of infected retroperitoneal fluid collections, such as infected pancreatic necrosis and perinephric abscess. Video-assisted retroperitoneal debridement (VARD) is a well-established approach in infected pancreatic necrosis when surgical debridement is warranted. Minimally invasive retroperitoneal approaches have emerged in a broadening range of etiologies and specialties. We describe our experience utilizing VARDs in two patients that developed retroperitoneal abscesses following gunshot injuries to bowel and proximal urinary system. Both failed a conservative approach including antibiotic and percutaneous drains. Rapid improvement and subsequent discharge were observed within days of VARD procedure. We believe VARD to be a viable approach to post-trauma retroperitoneal abscesses when surgical drainage is indicated, and anatomy is favorable.

2.
Hum Gene Ther ; 33(21-22): 1126-1141, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35994386

RESUMEN

The nonclinical safety assessments for gene therapies are evolving, leveraging over 20 years of experimental and clinical experience. Despite the growing experience with these therapeutics, there are no approved harmonized global regulatory documents for developing gene therapies with only the ICH (International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use) S12 guidance on nonclinical biodistribution currently under discussion. Several health authorities have issued guidance over the last 15 years on the nonclinical safety aspects for gene therapy products, but many of the recommendations are limited to high-level concepts on nonclinical safety aspects or altogether silent on key topics. Historically, this approach was appropriately vague given our relatively small dataset of nonclinical experience, where a comprehensive and detailed regulatory guidance approach was unlikely to be appropriate to address all scenarios. However, harmonization of key considerations and assumptions can provide a consistent basis for developing the appropriate nonclinical safety development plans for individual programs, reducing uncertainty across regulatory regions and unnecessary animal use. Several key areas of nonclinical safety testing are nearing maturation for a harmonized approach, including species selection, certain aspects of study design, study duration, and unintended genomic integration risks. Furthermore, several emerging topics are unaddressed in current regulatory guidance for gene therapy products, which will become key areas of differentiation for the next generation of therapeutics. These topics include redosing, juvenile/pediatric safety, and reproductive/developmental safety testing, where relevant experience from other modalities can be applied. The rationale and potential study design considerations for these topics will be proposed, acknowledging that certain aspects of gene therapy development are not considered appropriate for harmonization. This article provides an overview of the current nonclinical safety regulatory landscape, summarizes typical nonclinical safety study designs, highlights areas of uncertainty, and discusses emerging topics that warrant consideration. Specific recommendations and perspectives are provided to inform future regulatory discussions and harmonization efforts.


Asunto(s)
Terapia Genética , Humanos , Animales , Niño , Distribución Tisular , Terapia Genética/efectos adversos
3.
Regul Toxicol Pharmacol ; 127: 105064, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34656748

RESUMEN

Over the last decade, immunotherapy has established itself as an important novel approach in the treatment of cancer, resulting in a growing importance in oncology. Engineered T cell therapies, namely chimeric antigen receptor (CAR) T cells and T cell receptor (TCR) T cell therapies, are platform technologies that have enabled the development of products with remarkable efficacy in several hematological malignancies and are thus the focus of intense research and development activity. While engineered T cell therapies offer promise in addressing currently intractable cancers, they also present unique challenges, including their nonclinical safety assessment. A workshop organized by HESI and the US Food and Drug Administration (FDA) was held to provide an interdisciplinary forum for representatives of industry, academia and regulatory authorities to share information and debate on current practices for the nonclinical safety evaluation of engineered T cell therapies. This manuscript leverages what was discussed at this workshop to provide an overview of the current important nonclinical safety assessment considerations for the development of these therapeutic modalities (cytokine release syndrome, neurotoxicity, on-target/off-tumor toxicities, off-target effects, gene editing or vector integration-associated genomic injury). The manuscript also discusses approaches used for hazard identification or risk assessment and provides a regulatory perspective on such aspects.


Asunto(s)
Ingeniería Celular/métodos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Linfocitos T/inmunología , Síndrome de Liberación de Citoquinas/fisiopatología , Edición Génica , Inmunoterapia Adoptiva/efectos adversos , Síndromes de Neurotoxicidad/fisiopatología , Receptores de Antígenos de Linfocitos T/fisiología , Medición de Riesgo
4.
J Immunother ; 43(4): 107-120, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31899702

RESUMEN

Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment for patients with CD19 B-cell malignancies. Combination strategies that improve CAR T-cell potency, limit tumor environment-mediated immune dysfunction, and directly reduce tumor burden may increase the potential for durable clinical benefit of CAR T-cell therapy. Lisocabtagene maraleucel (liso-cel) is a product therapy candidate being tested in patients with relapsed/refractory non-Hodgkin lymphoma or chronic lymphocytic leukemia. This study assessed the in vitro and in vivo functionality of CAR T cells transduced to express the anti-CD19 CAR of liso-cel in combination with ibrutinib or acalabrutinib. In prolonged stimulation assays, the presence of ibrutinib or acalabrutinib improved the CAR T-cell effector function. RNA-Seq analysis and surface marker profiling of these CAR T cells treated with ibrutinib but not acalabrutinib revealed gene expression changes consistent with skewing toward a memory-like, type 1 T-helper, Bruton tyrosine kinase phenotype. Ibrutinib or acalabrutinib improved CD19 tumor clearance and prolonged survival of tumor-bearing mice when used in combination with CAR T cells. A combination of the defined cell product therapy candidate, liso-cel, with ibrutinib or acalabrutinib is an attractive approach that may potentiate the promising clinical responses already achieved in CD19 B-cell malignancies with each of these single agents.


Asunto(s)
Antígenos CD19/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Adenina/administración & dosificación , Adenina/análogos & derivados , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Benzamidas/administración & dosificación , Biomarcadores , Terapia Combinada , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Humanos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos/inmunología , Ratones , Neoplasias/etiología , Neoplasias/metabolismo , Piperidinas/administración & dosificación , Pirazinas/administración & dosificación , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Front Immunol ; 11: 577027, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33391257

RESUMEN

Chimeric antigen receptor (CAR) T cells provide new therapeutic options for patients with relapsed/refractory hematologic malignancies. However, neurotoxicity is a frequent, and potentially fatal, complication. The spectrum of manifestations ranges from delirium and language dysfunction to seizures, coma, and fatal cerebral edema. This novel syndrome has been designated immune effector cell-associated neurotoxicity syndrome (ICANS). In this review, we draw an arc from our current understanding of how systemic and potentially local cytokine release act on the CNS, toward possible preventive and therapeutic approaches. We systematically review reported correlations of secreted inflammatory mediators in the serum/plasma and cerebrospinal fluid with the risk of ICANS in patients receiving CAR T cell therapy. Possible pathophysiologic impacts on the CNS are covered in detail for the most promising candidate cytokines, including IL-1, IL-6, IL-15, and GM-CSF. To provide insight into possible final common pathways of CNS inflammation, we place ICANS into the context of other systemic inflammatory conditions that are associated with neurologic dysfunction, including sepsis-associated encephalopathy, cerebral malaria, thrombotic microangiopathy, CNS infections, and hepatic encephalopathy. We then review in detail what is known about systemic cytokine interaction with components of the neurovascular unit, including endothelial cells, pericytes, and astrocytes, and how microglia and neurons respond to systemic inflammatory challenges. Current therapeutic approaches, including corticosteroids and blockade of IL-1 and IL-6 signaling, are reviewed in the context of what is known about the role of cytokines in ICANS. Throughout, we point out gaps in knowledge and possible new approaches for the investigation of the mechanism, prevention, and treatment of ICANS.


Asunto(s)
Barrera Hematoencefálica/fisiología , Sistema Nervioso Central/inmunología , Citocinas/metabolismo , Neoplasias Hematológicas/terapia , Inmunoterapia Adoptiva/métodos , Inflamación/inmunología , Síndromes de Neurotoxicidad/metabolismo , Animales , Neoplasias Hematológicas/inmunología , Humanos , Síndromes de Neurotoxicidad/etiología , Receptores Quiméricos de Antígenos/genética
6.
Cancer Discov ; 8(6): 750-763, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29563103

RESUMEN

Chimeric antigen receptor (CAR) T-cell immunotherapy has revolutionized the treatment of refractory leukemias and lymphomas, but is associated with significant toxicities, namely cytokine release syndrome (CRS) and neurotoxicity. A major barrier to developing therapeutics to prevent CAR T cell-mediated neurotoxicity is the lack of clinically relevant models. Accordingly, we developed a rhesus macaque (RM) model of neurotoxicity via adoptive transfer of autologous CD20-specific CAR T cells. Following cyclophosphamide lymphodepletion, CD20 CAR T cells expand to 272 to 4,450 cells/µL after 7 to 8 days and elicit CRS and neurotoxicity. Toxicities are associated with elevated serum IL6, IL8, IL1RA, MIG, and I-TAC levels, and disproportionately high cerebrospinal fluid (CSF) IL6, IL2, GM-CSF, and VEGF levels. During neurotoxicity, both CD20 CAR and non-CAR T cells accumulate in the CSF and in the brain parenchyma. This RM model demonstrates that CAR T cell-mediated neurotoxicity is associated with proinflammatory CSF cytokines and a pan-T cell encephalitis.Significance: We provide the first immunologically relevant, nonhuman primate model of B cell-directed CAR T-cell therapy-mediated CRS and neurotoxicity. We demonstrate CAR and non-CAR T-cell infiltration in the CSF and in the brain during neurotoxicity resulting in pan-encephalitis, accompanied by increased levels of proinflammatory cytokines in the CSF. Cancer Discov; 8(6); 750-63. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 663.


Asunto(s)
Antígenos CD20/inmunología , Ciclofosfamida/administración & dosificación , Inmunoterapia Adoptiva/efectos adversos , Síndromes de Neurotoxicidad/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Ciclofosfamida/efectos adversos , Modelos Animales de Enfermedad , Humanos , Células K562 , Macaca mulatta , Síndromes de Neurotoxicidad/etiología , Trasplante Autólogo
7.
Clin Cancer Res ; 23(20): 6190-6202, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28706012

RESUMEN

Purpose: Talimogene laherparepvec, a new oncolytic immunotherapy, has been recently approved for the treatment of melanoma. Using a murine version of the virus, we characterized local and systemic antitumor immune responses driving efficacy in murine syngeneic models.Experimental Design: The activity of talimogene laherparepvec was characterized against melanoma cell lines using an in vitro viability assay. Efficacy of OncoVEXmGM-CSF (talimogene laherparepvec with the mouse granulocyte-macrophage colony-stimulating factor transgene) alone or in combination with checkpoint blockade was characterized in A20 and CT-26 contralateral murine tumor models. CD8+ depletion, adoptive T-cell transfers, and Enzyme-Linked ImmunoSpot assays were used to study the mechanism of action (MOA) of systemic immune responses.Results: Treatment with OncoVEXmGM-CSF cured all injected A20 tumors and half of contralateral tumors. Viral presence was limited to injected tumors and was not responsible for systemic efficacy. A significant increase in T cells (CD3+/CD8+) was observed in injected and contralateral tumors at 168 hours. Ex vivo analyses showed these cytotoxic T lymphocytes were tumor-specific. Increased neutrophils, monocytes, and chemokines were observed in injected tumors only. Importantly, depletion of CD8+ T cells abolished all systemic efficacy and significantly decreased local efficacy. In addition, immune cell transfer from OncoVEXmGM-CSF-cured mice significantly protected from tumor challenge. Finally, combination of OncoVEXmGM-CSF and checkpoint blockade resulted in increased tumor-specific CD8+ anti-AH1 T cells and systemic efficacy.Conclusions: The data support a dual MOA for OncoVEXmGM-CSF that involves direct oncolysis of injected tumors and activation of a CD8+-dependent systemic response that clears injected and contralateral tumors when combined with checkpoint inhibition. Clin Cancer Res; 23(20); 6190-202. ©2017 AACR.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunoterapia , Neoplasias/inmunología , Neoplasias/metabolismo , Viroterapia Oncolítica , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Adenoviridae/genética , Traslado Adoptivo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Inmunomodulación , Inmunoterapia/métodos , Estimación de Kaplan-Meier , Depleción Linfocítica , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Melanoma/terapia , Ratones , Neoplasias/patología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Transgenes , Carga Tumoral , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Arch Sex Behav ; 44(7): 2041-53, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25896489

RESUMEN

This qualitative study explored the social experiences and HIV-related sexual practices of 30 young gay and bisexual men who moved to New York City in the past 3 years from other countries or elsewhere in the United States. For many migrants, a key basis of vulnerability to HIV was their engagement with New York City's unfamiliar sexual culture. Many recent arrivals migrated from places with small gay communities and low HIV prevalence, and some came with a practice of limited condom use. Participants described encountering an abundance of sexual opportunity in New York City, accessible to even the newest arrivals through internet sex sites. Some migrants expressed surprise that few men they met were interested in dating or establishing trust before having sex. Although frequent HIV testing was common, HIV status, testing history, and condom use were seldom discussed with sex partners for some men even with new partners or before sex without condoms. International and in-country migrants who are beginning to navigate New York City's gay sexual culture may be more vulnerable to HIV infection than established residents if they are inexperienced in encountering vast sexual opportunity, are less practiced in local norms of sexual communication, or if their lack of economic resources or social connections encourages them to have sex for money or shelter. This article suggests HIV prevention interventions for urban migrants and other men who have sex with men.


Asunto(s)
Infecciones por VIH/etiología , Homosexualidad Masculina/psicología , Síndrome de Inmunodeficiencia Adquirida , Adolescente , Adulto , Humanos , Masculino , Ciudad de Nueva York/epidemiología , Conducta de Reducción del Riesgo , Conducta Sexual , Parejas Sexuales , Estados Unidos , Adulto Joven
9.
Curr Med Res Opin ; 31(3): 557-74, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25651481

RESUMEN

OBJECTIVE: Tumor necrosis factor (TNF) is a highly pleiotropic cytokine with multiple activities other than its originally discovered role of tumor necrosis in rodents. TNF is now understood to play a contextual role in driving either tumor elimination or promotion. Using both animal and human data, this review examines the role of TNF in cancer development and the effect of TNF and TNF inhibitors (TNFis) on malignancy risk. RESEARCH DESIGN: A literature review was performed using relevant search terms for TNF and malignancy. RESULTS: Although administration of TNF can cause tumor regression in specific rodent tumor models, human expression polymorphisms suggest that TNF can be a tumor-promoting cytokine, whereas blocking the TNF pathway in a variety of tumor models inhibits tumor growth. In addition to direct effects of TNF on tumors, TNF can variously affect immunity and the tumor microenvironment. Whereas TNF can promote immune surveillance designed to eliminate tumors, it can also drive chronic inflammation, autoimmunity, angiogenesis, and other processes that promote tumor initiation, growth, and spread. Key players in TNF signaling that shape this response include NF-κB and JNK, and malignant-inflammatory cell interactions, each of which may have different responses to TNF signaling. Focusing on rheumatoid arthritis (RA) patients, where clinical experience is most extensive, a review of the clinical literature shows no increased risk of overall malignancy or solid tumors such as breast and lung cancers with exposure to TNFis. Lymphoma rates are not increased with use of TNFis. Conflicting data exist regarding the risks of melanoma and nonmelanoma skin cancer. Data regarding the risk of recurrent malignancy are limited. CONCLUSIONS: Overall, the available data indicate that elevated TNF is a risk factor for cancer, whereas its inhibition in RA patients is not generally associated with an increased cancer risk. In particular, TNF inhibition is not associated with cancers linked to immune suppression. A better understanding of the tumor microenvironment, molecular events underlying specific tumors, and epidemiologic studies of malignancies within specific disease indications should enable more focused pharmacovigilance studies and a better understanding of the potential risks of TNFis.


Asunto(s)
Artritis Reumatoide/inmunología , Inflamación/inmunología , Neoplasias , Factores de Necrosis Tumoral , Animales , Modelos Animales de Enfermedad , Humanos , Recurrencia Local de Neoplasia/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Factores de Riesgo , Transducción de Señal , Microambiente Tumoral , Inhibidores del Factor de Necrosis Tumoral , Factores de Necrosis Tumoral/metabolismo
10.
Mycorrhiza ; 24 Suppl 1: S55-64, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24487451

RESUMEN

Although the important effects of pH and carbonate content of soils on "black truffle" (Tuber melanosporum) production are well known, we poorly understand the influence of soil physical properties. This study focuses on physical soil characteristics that drive successful production of black truffles in plantations. Seventy-eight Quercus ilex ssp. ballota plantations older than 10 years were studied in the province of Teruel (eastern Spain). Soil samples were analyzed for various edaphic characteristics and to locate T. melanosporum ectomycorrhizae. The influence of cultivation practices, climatic features, and soil properties on sporocarp production was assessed using multivariate analyses. Low contents of fine earth and silt and high levels of bulk density, clay content, and water-holding capacity appear to promote fructification. Watering is also highly positive for truffle fructification. We develop and discuss a logistic model to predict the probability of truffle fructification in field sites under consideration for truffle plantation establishment. The balance between water availability and aeration plays a crucial role in achieving success in black truffle plantations.


Asunto(s)
Ascomicetos/fisiología , Cuerpos Fructíferos de los Hongos/fisiología , Micorrizas/fisiología , Suelo/química , Agricultura , Ascomicetos/crecimiento & desarrollo , Cuerpos Fructíferos de los Hongos/crecimiento & desarrollo , Modelos Logísticos , Análisis Multivariante , Micorrizas/crecimiento & desarrollo , Quercus/microbiología , Microbiología del Suelo
11.
Oncolytic Virother ; 3: 11-20, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-27512660

RESUMEN

Oncolytic viruses that selectively lyse tumor cells with minimal damage to normal cells are a new area of therapeutic development in oncology. An attenuated herpesvirus encoding the granulocyte-macrophage colony stimulating factor (GM-CSF), known as talimogene laherparepvec (T-VEC), has been identified as an attractive oncolytic virus for cancer therapy based on preclinical tumor studies and results from early-phase clinical trials and a large randomized Phase III study in melanoma. In this review, we discuss the basic biology of T-VEC, describe the role of GM-CSF as an immune adjuvant, summarize the preclinical data, and report the outcomes of published clinical trials using T-VEC. The emerging data suggest that T-VEC is a safe and potentially effective antitumor therapy in malignant melanoma and represents the first oncolytic virus to demonstrate therapeutic activity against human cancer in a randomized, controlled Phase III study.

12.
J Immunotoxicol ; 11(1): 1-12, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23746314

RESUMEN

Observational and clinical studies have associated increased cancer risks with primary or acquired immunodeficiencies, autoimmunity, and use of immunotherapies to treat chronic inflammation (e.g. autoimmunity) or support organ engraftment. Understanding of the relationship between immune status and cancer risk is generally grounded in two juxtaposing paradigms: that the immune system protects the host via surveillance of tumors and oncogenic viruses (e.g. immunosurveillance model) and that chronic inflammation can augment tumor growth and metastasis (inflammation model). Whereas these models support a role of immune status in many cancers, they are insufficient to explain the disproportionate increase in B-cell lymphoma risk observed across patient populations with either chronic immunosuppression or inflammation. Evaluation for the presence of Epstein-Barr virus (EBV) in lymphomas obtained from various populations demonstrates a variable role for the virus in lymphomagenesis across patient populations. An evaluation of the DNA alterations found in lymphomas and an understanding of B-cell ontogeny help to provide insight into the unique susceptibility of lymphocytes, primarily B-cells, to oncogenic transformation. EBV-independent B-cell oncogenic transformation is driven by chronic antigenic stimulation due to either inflammation (as seen in patients with autoimmune disease or a tissue allograft) or to unresolved infection (as seen in immunosuppressed patients), and the transformation arises as a result of DNA damage from genomic recombination and mutation during class switching and somatic hypermutation. This model explains the increased background rate of lymphoma in some patients with autoimmunity, and highlights the challenge of resolving the confounding that occurs between disease severity and use of targeted immunotherapies to treat chronic inflammation. The ability to distinguish between disease- and treatment-related risk of lymphoma and an appreciation of the etiology of B-cell transformation is central to an improved risk assessment by scientists, clinicians and regulators, including the approval, labeling, and chronic use of immunotherapies.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Virus de Epstein-Barr/terapia , Herpesvirus Humano 4/inmunología , Inmunoterapia/métodos , Linfoma de Células B/terapia , Animales , Linfocitos B/virología , Transformación Celular Neoplásica , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Humanos , Huésped Inmunocomprometido , Inmunomodulación , Linfoma de Células B/etiología , Linfoma de Células B/inmunología , Medición de Riesgo
13.
PLoS One ; 8(6): e67256, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825648

RESUMEN

Rituximab, a monoclonal antibody targeting CD20 on B cells, is currently used to treat many subtypes of B cell lymphomas. However, treatment is not curative and response rates are variable. Recombinant interleukin-21 (rIL-21) is a cytokine that enhances immune effector function and affects both primary and transformed B cell differentiation. We hypothesized that the combination of rIL-21 plus rituximab would be a more efficacious treatment for B cell malignancies than rituximab alone. We cultured human and cynomolgus monkey NK cells with rIL-21 and found that their activity was increased and proteins associated with antibody dependent cytotoxicity were up-regulated. Studies in cynomolgus monkeys modeled the effects of rIL-21 on rituximab activity against CD20 B cells. In these studies, rIL-21 activated innate immune effectors, increased ADCC and mobilized B cells into peripheral blood. When rIL-21 was combined with rituximab, deeper and more durable B cell depletion was observed. In another series of experiments, IL-21 was shown to have direct antiproliferative activity against a subset of human lymphoma cell lines, and combination of murine IL-21 with rituximab yielded significant survival benefits over either agent alone in xenogeneic mouse tumor models of disseminated lymphoma. Therefore, our results do suggest that the therapeutic efficacy of rituximab may be improved when used in combination with rIL-21.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Antineoplásicos/farmacología , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Interleucinas/farmacología , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antineoplásicos/uso terapéutico , Linfocitos B/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Linfoma de Células B/patología , Macaca fascicularis , Masculino , Ratones , Rituximab , Análisis de Supervivencia
14.
Int J Toxicol ; 31(4): 303-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22723515

RESUMEN

Interleukin-21 (IL-21), a pleiotropic immunostimulatory type I cytokine, has anticancer effects in animal models. Preclinical studies designed to assess the safety of recombinant human IL-21 (rIL-21) for use in phase I oncology studies are described. The rIL-21 (≤3.0 mg/kg per dose) was given intravenously to cynomolgus monkeys (Macaca fascicularis) once daily for 5 days, followed by 9 nondosing days (1 cycle) for ≤4 cycles. The rIL-21 pharmacokinetics was dose-dependent. Accumulation was not observed after repeated dosing, consistent with the short elimination half-life (t (1/2,λz); 0.4-0.8 hours). Safety findings included cyclical anemia and thrombocytopenia, clinical pathology changes consistent with acute-phase response, leukocyte infiltrates in hepatic sinusoids, and sporadic serum transaminase elevations (typically <3 times upper-limit of normal); all were reversible upon cessation of treatment. Decreased pharmacodynamic responses with time corresponded to the development of anti-rIL-21 antibodies; effects varied among individuals and were dose-dependent. These studies demonstrated rIL-21 to be generally well-tolerated when administered to cynomolgus monkeys, and all adverse effects were reversible upon treatment cessation. However, development of anti-rIL-21 antibodies may limit the use of this species in long-term studies.


Asunto(s)
Interleucinas/farmacología , Interleucinas/farmacocinética , Reacción de Fase Aguda/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Semivida , Humanos , Interleucinas/sangre , Macaca fascicularis , Masculino , Fosforilación , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT3/metabolismo
15.
J Immunotoxicol ; 8(4): 389-97, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21913866

RESUMEN

Regulatory T-cell (T(reg)) modulation is developing as an important therapeutic opportunity for the treatment of a number of important diseases, including cancer, autoimmunity, infection, and organ transplant rejection. However, as demonstrated with IL-2 and TGN-1412, our understanding of the complex immunological interactions that occur with T(reg) modulation in both non-clinical models and in patients remains limited and appears highly contextual. This lack of understanding will challenge our ability to identify the patient population who might derive the highest benefit from T(reg) modulation and creates special challenges as we transition these therapeutics from non-clinical models into humans. Thus, in vivo testing in the most representative animal model systems, with careful progress in the clinic, will remain critical in developing therapeutics targeting T(reg) and understanding their clinical utility. Moreover, toxicology models can inform some of the potential liabilities associated with T(reg) modulation, but not all, suggesting a continued need to explore and validate predictive models.


Asunto(s)
Productos Biológicos/efectos adversos , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Factores Inmunológicos/efectos adversos , Linfocitos T Reguladores/efectos de los fármacos , Animales , Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos/efectos adversos , Humanos , Interleucina-2/efectos adversos , Medición de Riesgo , Linfocitos T Reguladores/inmunología
16.
J Immunotoxicol ; 8(4): 251-7, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21848365

RESUMEN

In addition to the effector T-cells subsets, T-cells can also differentiate into cells that play a suppressive or regulatory role in adaptive immune responses. The cell types currently identified as regulatory T-cells (T(regs)) include natural or thymic-derived T(regs), T-cells which express Foxp3(+)CD25(+)CD4(+) and can suppress immune responses to autoreactive T-cells, as well as inducible T(regs), that are generated from naïve T-cells in the periphery after interaction with antigens presented by dendritic cells. Inducible T(regs) include T(H)3 cells, T(r)1 cells, and Foxp3(+)-inducible T(regs). T(regs) have been shown to be critical in the maintenance of immune responses and T-cell homeostasis. These cells play an important role in suppressing responses to self-antigens and in controlling inappropriate responses to non-self-antigens, such as commensal bacteria or food in the gut. For example, depletion of CD4(+)CD25(+) T(regs) from mice resulted in the development of multi-organ autoimmune diseases. CD4(+)CD25(+) T(regs) and/or IL-10-producing T(r)1 cells are capable of suppressing or attenuating T(H)2 responses to allergens. Moreover, adoptive transfer of CD4(+)CD25(+) T(regs) from healthy to diseased animals resulted in the prevention or cure of certain autoimmune diseases, and was able to induce transplantation tolerance. Clinical improvement seen after allergen immunotherapy for allergic diseases such as rhinitis and asthma is associated with the induction of IL-10- and TGFß-producing T(r)1 cells as well as FoxP3-expressing IL-10 T-cells, with resulting suppression of the T(H)2 cytokine milieu. Activation, expansion, or suppression of CD4(+)CD25(+) T(regs) in vivo by xenobiotics, including drugs, may therefore represent a relevant mechanism underlying immunotoxicity, including immunosuppression, allergic asthma, and autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inducido químicamente , Contaminantes Ambientales/toxicidad , Hipersensibilidad/inmunología , Inmunidad Celular/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Linfocitos T Reguladores/inmunología , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Autoantígenos , Biomarcadores/metabolismo , Células Dendríticas/inmunología , Cobayas , Inmunidad Celular/inmunología , Terapia de Inmunosupresión , Ratones , Ratas , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
17.
Toxicol Pathol ; 39(6): 913-5, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21859887

RESUMEN

International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) S9 is the first international regulatory guidance document devoted to a specific therapeutic area, highlighting its importance in harmonizing regulatory expectations for the nonclinical development of therapeutics designed to treat advanced cancer. ICH S9 successfully outlines the core requirements for the nonclinical development of novel oncology therapeutics, and the unique risk-benefit considerations for expediting drug development to treat these most grievous diseases. However, development companies and regulatory agencies have had limited opportunity to apply this guidance, so our collective experience in developing therapeutics under the guidance is limited. Discussed here are several key areas of ambiguity in the guidance, including identification of the patient population, selection of the initial safe dose for first-in-human trials, and requirements for nonclinical recovery data.


Asunto(s)
Anticarcinógenos/síntesis química , Neoplasias/tratamiento farmacológico , Preparaciones Farmacéuticas/síntesis química , Animales , Anticarcinógenos/efectos adversos , Relación Dosis-Respuesta a Droga , Aprobación de Drogas , Evaluación Preclínica de Medicamentos , Guías como Asunto , Humanos , Medición de Riesgo , Estados Unidos , United States Food and Drug Administration
18.
Toxicol Sci ; 120(2): 403-12, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21252392

RESUMEN

The tumor suppressor oncoprotein, p53, is a critical regulator of stress-induced growth arrest and apoptosis. p53 activity is regulated through the ubiquitin proteasome system (UPS) with stress-induced disruption leading to increased accumulation of p53, resulting in growth arrest. In the present study, we investigate the role of p53 to determine sensitivity to cadmium (Cd) and whether induction of stress signaling responses and perturbation of the UPS are involved in Cd-induced cytotoxicity and apoptosis. We treated synchronously cultured p53 transgenic mouse embryonic fibroblasts, both wild-type p53+/+ and knockout p53-/- cells, with cadmium chloride (Cd, 0.5-20µM) for 24 h. Cd-induced cytotoxicity was assessed by cellular morphology disruption and neutral red dye uptake assay. Proteins in the stress signaling pathway, including p38 mitogen-activated protein kinase (MAPK) and stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK); ubiquitination, such as high-molecular weight of polyubiquitinated proteins (HMW-polyUb); and apoptotic pathways, were all measured. We found that Cd induced p53-dependent cytotoxicity in the p53+/+ cells, which exhibited a twofold greater sensitivity. We observed a dose-dependent stimulation of p38 MAPK and SAPK/JNK phosphorylation that corresponded to accumulation of HMW-polyUb conjugates and lead to the induction of apoptosis, as evidenced by the elevation of cleaved caspase-3. Our study suggests that Cd-mediated cytotoxicity and induction of stress signaling responses, elevated accumulation of HMW-polyUb conjugates, and resulting apoptosis are all dependent on p53 status.


Asunto(s)
Apoptosis/efectos de los fármacos , Cloruro de Cadmio/toxicidad , Contaminantes Ambientales/toxicidad , Fibroblastos/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Proteína p53 Supresora de Tumor/fisiología , Proteínas Ubiquitinadas/metabolismo , Animales , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Inmunoprecipitación , Ratones , Ratones Noqueados , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína p53 Supresora de Tumor/genética
19.
Toxicol Pathol ; 38(4): 522-53, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20472697

RESUMEN

An important safety consideration for developing new therapeutics is assessing the potential that the therapy will increase the risk of cancer. For biotherapeutics, traditional two-year rodent bioassays are often not scientifically applicable or feasible. This paper is a collaborative effort of industry toxicologists to review past and current practice regarding carcinogenicity assessments of biotherapeutics and to provide recommendations. Publicly available information on eighty marketed protein biotherapeutics was reviewed. In this review, no assessments related to carcinogenicity or tumor growth promotion were identified for fifty-one of the eighty molecules. For the twenty-nine biotherapeutics in which assessments related to carcinogenicity were identified, various experimental approaches were employed. This review also discusses several key principles to aid in the assessment of carcinogenic potential, including (1) careful consideration of mechanism of action to identify theoretical risks, (2) careful investigation of existing data for indications of proliferative or immunosuppressive potential, and (3) characterization of any proliferative or immunosuppressive signals detected. Traditional two-year carcinogenicity assays should not be considered as the default method for assessing the carcinogenicity potential of biotherapeutics. If experimentation is considered warranted, it should be hypothesis driven and may include a variety of experimental models. Ultimately, it is important that preclinical data provide useful guidance in product labeling.


Asunto(s)
Biofarmacia/métodos , Biotecnología/métodos , Pruebas de Carcinogenicidad/métodos , Aprobación de Drogas/métodos , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos
20.
Regul Toxicol Pharmacol ; 54(2): 164-82, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19345250

RESUMEN

An evaluation of potential antibody formation to biologic therapeutics during the course of nonclinical safety studies and its impact on the toxicity profile is expected under current regulatory guidance and is accepted standard practice. However, approaches for incorporating this information in the interpretation of nonclinical safety studies are not clearly established. Described here are the immunological basis of anti-drug antibody formation to biopharmaceuticals (immunogenicity) in laboratory animals, and approaches for generating and interpreting immunogenicity data from nonclinical safety studies of biotechnology-derived therapeutics to support their progression to clinical evaluation. We subscribe that immunogenicity testing strategies should be adapted to the specific needs of each therapeutic development program, and data generated from such analyses should be integrated with available clinical and anatomic pathology, pharmacokinetic, and pharmacodynamic data to properly interpret nonclinical studies.


Asunto(s)
Formación de Anticuerpos/efectos de los fármacos , Biofarmacia/métodos , Proteínas Recombinantes/toxicidad , Pruebas de Toxicidad/métodos , Animales , Biofarmacia/estadística & datos numéricos , Interpretación Estadística de Datos , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Humanos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacocinética , Especificidad de la Especie , Pruebas de Toxicidad/estadística & datos numéricos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...