Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Clin Pharmacol Ther ; 112(4): 882-891, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35694844

RESUMEN

With the ongoing global pandemic of coronavirus disease 2019 (COVID-19), there is an urgent need to accelerate the traditional drug development process. Many studies identified potential COVID-19 therapies based on promising nonclinical data. However, the poor translatability from nonclinical to clinical settings has led to failures of many of these drug candidates in the clinical phase. In this study, we propose a mechanism-based, quantitative framework to translate nonclinical findings to clinical outcome. Adopting a modularized approach, this framework includes an in silico disease model for COVID-19 (virus infection and human immune responses) and a pharmacological component for COVID-19 therapies. The disease model was able to reproduce important longitudinal clinical data for patients with mild and severe COVID-19, including viral titer, key immunological cytokines, antibody responses, and time courses of lymphopenia. Using remdesivir as a proof-of-concept example of model development for the pharmacological component, we developed a pharmacological model that describes the conversion of intravenously administered remdesivir as a prodrug to its active metabolite nucleoside triphosphate through intracellular metabolism and connected it to the COVID-19 disease model. After being calibrated with the placebo arm data, our model was independently and quantitatively able to predict the primary endpoint (time to recovery) of the remdesivir clinical study, Adaptive Covid-19 Clinical Trial (ACTT). Our work demonstrates the possibility of quantitatively predicting clinical outcome based on nonclinical data and mechanistic understanding of the disease and provides a modularized framework to aid in candidate drug selection and clinical trial design for COVID-19 therapeutics.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , Antivirales/uso terapéutico , Calibración , Humanos , Farmacología en Red , SARS-CoV-2
2.
Front Med (Lausanne) ; 9: 1109541, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36743666

RESUMEN

The U.S. Food and Drug Administration (FDA) Division of Applied Regulatory Science (DARS) moves new science into the drug review process and addresses emergent regulatory and public health questions for the Agency. By forming interdisciplinary teams, DARS conducts mission-critical research to provide answers to scientific questions and solutions to regulatory challenges. Staffed by experts across the translational research spectrum, DARS forms synergies by pulling together scientists and experts from diverse backgrounds to collaborate in tackling some of the most complex challenges facing FDA. This includes (but is not limited to) assessing the systemic absorption of sunscreens, evaluating whether certain drugs can convert to carcinogens in people, studying drug interactions with opioids, optimizing opioid antagonist dosing in community settings, removing barriers to biosimilar and generic drug development, and advancing therapeutic development for rare diseases. FDA tasks DARS with wide ranging issues that encompass regulatory science; DARS, in turn, helps the Agency solve these challenges. The impact of DARS research is felt by patients, the pharmaceutical industry, and fellow regulators. This article reviews applied research projects and initiatives led by DARS and conducts a deeper dive into select examples illustrating the impactful work of the Division.

3.
EMBO Mol Med ; 12(7): e8662, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32578942

RESUMEN

Mice xenotransplanted with human cells and/or expressing human gene products (also known as "humanized mice") recapitulate the human evolutionary specialization and diversity of genotypic and phenotypic traits. These models can provide a relevant in vivo context for understanding of human-specific physiology and pathologies. Humanized mice have advanced toward mainstream preclinical models and are now at the forefront of biomedical research. Here, we considered innovations and challenges regarding the reconstitution of human immunity and human tissues, modeling of human infections and cancer, and the use of humanized mice for testing drugs or regenerative therapy products. As the number of publications exploring different facets of humanized mouse models has steadily increased in past years, it is becoming evident that standardized reporting is needed in the field. Therefore, an international community-driven resource called "Minimal Information for Standardization of Humanized Mice" (MISHUM) has been created for the purpose of enhancing rigor and reproducibility of studies in the field. Within MISHUM, we propose comprehensive guidelines for reporting critical information generated using humanized mice.


Asunto(s)
Modelos Animales de Enfermedad , Guías como Asunto , Xenoinjertos/normas , Animales , Humanos , Ratones , Ratones SCID , Neoplasias , Reproducibilidad de los Resultados
4.
Sci Rep ; 10(1): 2476, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-32051479

RESUMEN

PEGylated recombinant human granulocyte colony stimulating factor (pegfilgrastim) is used clinically to accelerate immune reconstitution following chemotherapy and is being pursued for biosimilar development. One challenge to overcome in pegfilgrastim biosimilar development is establishing pharmacokinetic (PK) similarity, which is partly due to the degree of PK variability. We herein report that commercially available G-CSF and PEG ELISA detection kits have different capacities to detect pegfilgrastim aggregates that rapidly form in vitro in physiological conditions. These aggregates can be observed using SDS-PAGE, size-exclusion chromatography, dynamic light scattering, and real-time NMR analysis and are associated with decreased bioactivity as reflected by reduced drug-induced cellular proliferation and STAT3 phosphorylation. Furthermore, individual variability in the stability and detectability of pegfilgrastim in human sera is also observed. Pegfilgrastim levels display marked subject variability in sera from healthy donors incubated at 37 °C. The stability patterns of pegfilgrastim closely match the stability patterns of filgrastim, consistent with a key role for pegfilgrastim's G-CSF moiety in driving formation of inactive aggregates. Taken together, our results indicate that individual variability and ELISA specificity for inactive aggregates are key factors to consider when designing and interpreting studies involving the measurement of serum pegfilgrastim concentrations.


Asunto(s)
Variación Biológica Individual , Filgrastim/farmacocinética , Polietilenglicoles/farmacocinética , Animales , Línea Celular Tumoral , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática/normas , Humanos , Ratones , Factor de Transcripción STAT3/metabolismo
5.
Transl Res ; 210: 43-56, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31082370

RESUMEN

Cytokine release syndrome (CRS) is a serious and potentially life-threatening complication that can be associated with biological drug products. In vitro assays or in vivo tests using nonhuman primates may fail to predict CRS due to species differences and the complexity of immune system. Therefore, model species that have human-specific immune components may improve the ability to identify CRS and enhance product safety. In this study we used bone marrow-liver-thymus (BLT) humanized mice to test muromonab (OKT3), an anti-CD3 antibody with a black box warning for CRS. Initially, we completed pilot and dose escalation studies with muromonab and showed that when the dose was increased sufficiently, BLT-humanized mice experienced serious adverse outcomes including moribundity. Full studies compared muromonab treatment with adalimumab, saline, and a group pretreated with methylprednisolone prior to muromonab. We evaluated immune cell activation using flow cytometry and cytokine expression using a custom 10-plex cytokine assay to assess levels of human TNF-α, IFN-γ, IL-2, IL-6, IL-8, IL-10, IL-13, IL-17A, IL12/23p40, and GM-CSF. Muromonab treated mice had significant increases in all cytokines tested with T-cell depletion and T-cell activation noted. Adalimumab (active) and saline (inactive) control groups did not demonstrate cytokine expression changes or alterations in T-cell numbers or activation. Further, pretreatment with methylprednisolone blunted or abrogated cytokine increases. This study demonstrates that BLT-humanized mice are capable of experiencing CRS, and could be used to screen biologics for this adverse event to enhance patient safety.


Asunto(s)
Médula Ósea/inmunología , Citocinas/metabolismo , Hígado/inmunología , Timo/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Hígado/citología , Activación de Linfocitos/efectos de los fármacos , Recuento de Linfocitos , Ratones , Muromonab-CD3/farmacología , Bazo/citología , Síndrome , Linfocitos T/efectos de los fármacos
6.
Toxicol Sci ; 169(1): 194-208, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30850839

RESUMEN

Checkpoint inhibitors represent a new class of therapeutics in the treatment of cancer that has demonstrated remarkable clinical effectiveness. However, some patients have experienced serious immune-mediated adverse effects including pneumonitis, hepatitis, colitis, nephritis, dermatitis, encephalitis, and adrenal or pituitary insufficiency. These adverse events were not predicted by nonclinical studies. To determine if bone marrow-liver-thymus (BLT) immune humanized mice could demonstrate these adverse effects, we studied the effect of nivolumab on 2 strains of BLT-humanized mice, NOD.Cg-Prkdcscid Il2rgtm1Sug/JicTac (NOG) and NOD.Cg-Prkdcscid Il2rgtm1Sug Tg(SV40/HTLV-IL3, CSF2)10-7Jic/JicTac (NOG-EXL). Mice were treated with 2.5, 5.0, or 10.0 mg/kg nivolumab or saline twice weekly for 28 days. BLT-NOG mice had significantly reduced survival compared with BLT-NOG-EXL mice. In spite of the difference in survival, both BLT-humanized strains showed adverse reactions similar to those reported in humans, including pneumonitis and hepatitis, with nephritis, dermatitis and adrenalitis also noted in some individuals. Additional histopathologic findings included pancreatic atrophy, myositis, and osteomyelitis in some animals. T-cell activation increased with concomitant loss of PD-1 detection. These findings show that BLT immune humanized mice can demonstrate immune-mediated adverse effects of antiPD1 therapy, and may represent a model that can be used to better understand toxicity of this class of drugs.


Asunto(s)
Antineoplásicos Inmunológicos/toxicidad , Sistema Inmunológico/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Nivolumab/toxicidad , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Animales , Trasplante de Médula Ósea , Femenino , Genotipo , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Sistema Inmunológico/patología , Trasplante de Hígado , Ratones Endogámicos NOD , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal , Especificidad de la Especie , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Timo/inmunología , Timo/trasplante
7.
Toxicol Appl Pharmacol ; 372: 57-69, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30914376

RESUMEN

Cytokine release syndrome (CRS) is a serious and potentially life-threatening complication typically associated with biological drug products. Pre-clinical testing in vitro and in vivo studies using non-human primates had failed to reliably predict CRS. To determine if bone marrow-thymus-liver (BLT) humanized mice with a fully engrafted human immune system or a CD34-humanized mouse model could predict CRS, we tested an anti-CD28 monoclonal antibody (mAb) similar to TGN1412. This TGN1412 analogue (TGN1412A) was initially tested in vitro and found to produce significant dose-dependent increases in cytokine production. For in vivo studies, adalimumab, an anti-tumor necrosis factor-alpha antibody known not to cause CRS, served as a negative control. We evaluated immune cell activation and cytokine expression in three independent experiments. In BLT humanized mice, significant increases in levels of human cytokines were identified in animals treated with anti-CD28 mAb. As expected, CD28+ cell detection was strongly reduced in the anti-CD28 treated group. Increased T cell activation was also observed. The control group did not show reductions in CD28+ T-cells and did not experience increased cytokine levels. Responses by CD34-humanized mice showed no significant differences between adalimumab and anti-CD28 treatment at doses used to test BLT-humanized mice. These results suggest that the TGN1412A produces similar results in vitro to the original TGN1412 monoclonal antibody. The BLT immune humanized mice but not the CD34 humanized mice produce both robust and specific cytokine responses and may represent a pre-clinical model to identify CRS.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Antígenos CD28/antagonistas & inhibidores , Síndrome de Liberación de Citoquinas/etiología , Citocinas/sangre , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD34/inmunología , Antígenos CD28/sangre , Antígenos CD28/inmunología , Células Cultivadas , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/inmunología , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Trasplante de Hígado , Ratones Endogámicos NOD , Ratones SCID , Medición de Riesgo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Timo/embriología , Timo/trasplante
8.
Clin Transl Sci ; 12(3): 283-290, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30737892

RESUMEN

CD20 monoclonal antibodies are well-established therapeutics for the treatment of B-cell malignancies. Several mechanisms of target cell killing occur from anti-CD20 therapy, including complement-dependent cytotoxicity (CDC) cell lysis and antibody-dependent cell-mediated cytotoxicity. Human Fc receptors (FcRs) are required to mediate these functions and are either not present or not cross-reactive in mice and most animal species. In contrast, some nonhuman primates have cross-reactive FcR; however, their cellular expression and function may differ from humans. Therefore, we tested bone marrow-liver-thymus (BLT) humanized mice to determine if they could recapitulate the pharmacokinetics (PKs), pharmacodynamics, and potential toxicities of ofatumumab, for which CDC is the predominant mechanism of action. Ofatumumab-treated BLT mice depleted B cells in a dose-dependent manner in all tissues sampled and recapitulated the PKs observed in humans, suggesting that BLT mice can mediate the CDC effector mechanism associated with biological drug products.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Antígenos CD20/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Linfocitos B/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Humanos , Depleción Linfocítica , Ratones Endogámicos NOD
9.
Comp Med ; 68(5): 353-359, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30208988

RESUMEN

Unexpected mortality occurred in a group of 12 NOD.Cg-NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) and 12 NOD.Cg-Rag1tm1Mom Il2rgtm1Wjl/SzJ (NRG) immunodeficient mice. At 10 d after routine bone marrow-liver-thymus humanization surgery, 9 mice were found dead without observation of initiating clinical signs; 1 d later (day 11), 3 additional mice showed signs of morbidity, including severe hunching, lateral recumbency, slow movement, shallow respiration, and decreased response to external stimulus. All remaining mice rapidly decompensated and were found dead or were euthanized within 4 d after the first death. Histopathology revealed severe ascending pyelonephritis with numerous yeast. Cultures in some mice were positive for Enterococcus faecalis or Staphylococcus xylosus, 2 bacteria considered commensals in rodents. In addition, Candida albicans was cultured from some animals. Further investigation revealed that a restraining device used for tail vein injections was the likely fomite harboring Candida organisms. These findings indicate that ascending pyelonephritis, with Candida as the etiologic agent, can cause significant mortality in NSG and NRG immunodeficient mice.


Asunto(s)
Candidiasis/veterinaria , Infecciones Oportunistas/veterinaria , Pielonefritis/veterinaria , Enfermedades de los Roedores/microbiología , Animales , Candidiasis/complicaciones , Candidiasis/epidemiología , Brotes de Enfermedades/veterinaria , Femenino , Humanos , Huésped Inmunocomprometido , Ratones Endogámicos NOD , Infecciones Oportunistas/complicaciones , Pielonefritis/complicaciones , Pielonefritis/epidemiología , Pielonefritis/microbiología , Enfermedades de los Roedores/epidemiología , Trasplante de Tejidos
10.
Part Fibre Toxicol ; 14(1): 25, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28716104

RESUMEN

BACKGROUND: As nanoparticles (NPs) become more prevalent in the pharmaceutical industry, questions have arisen from both industry and regulatory stakeholders about the long term effects of these materials. This study was designed to evaluate whether gold (10 nm), silver (50 nm), or silica (10 nm) nanoparticles administered intravenously to mice for up to 8 weeks at doses known to be sub-toxic (non-toxic at single acute or repeat dosing levels) and clinically relevant could produce significant bioaccumulation in liver and spleen macrophages. RESULTS: Repeated dosing with gold, silver, and silica nanoparticles did not saturate bioaccumulation in liver or spleen macrophages. While no toxicity was observed with gold and silver nanoparticles throughout the 8 week experiment, some effects including histopathological and serum chemistry changes were observed with silica nanoparticles starting at week 3. No major changes in the splenocyte population were observed during the study for any of the nanoparticles tested. CONCLUSIONS: The clinical impact of these changes is unclear but suggests that the mononuclear phagocytic system is able to handle repeated doses of nanoparticles.


Asunto(s)
Oro/toxicidad , Hígado/efectos de los fármacos , Macrófagos/efectos de los fármacos , Nanopartículas , Dióxido de Silicio/toxicidad , Plata/toxicidad , Bazo/efectos de los fármacos , Animales , Biomarcadores/sangre , Femenino , Oro/administración & dosificación , Oro/metabolismo , Inyecciones Intravenosas , Hígado/metabolismo , Hígado/patología , Macrófagos/metabolismo , Macrófagos/patología , Nanopartículas del Metal , Ratones Endogámicos BALB C , Medición de Riesgo , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/metabolismo , Plata/administración & dosificación , Plata/metabolismo , Bazo/metabolismo , Bazo/patología , Factores de Tiempo , Distribución Tisular
11.
Toxicol Appl Pharmacol ; 287(3): 246-52, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26079829

RESUMEN

The erythropoietin analog peginesatide was withdrawn from marketing due to unexpected severe anaphylactic reactions associated with administration of the multi-use formulation. The adverse events occurred rapidly following the first ever administration of the drug with most affected patients becoming symptomatic in less than 30min. This is most consistent with an anaphylactoid reaction due to direct activation of mast cells. Laboratory evaluation was undertaken using rat peritoneal mast cells as the model system. Initial studies showed that high concentrations of the formulated drug as well as formulated vehicle alone could cause mast cell degranulation as measured by histamine release. The purified active drug was not able to cause histamine release whereas the vehicle filtrate and lab created drug vehicle were equally potent at causing histamine release. Individual formulations of vehicle leaving one component out showed that histamine release was due to phenol. Dose response studies with phenol showed a very sharp dose response curve that was similar in three buffer systems. Cellular analysis by flow cytometry showed that the histamine release was not due to cell death, and that changes in light scatter parameters consistent with degranulation were rapidly observed. Limited testing with primary human mast cells showed a similar dose response of histamine release with exposure to phenol. To provide in vivo confirmation, rats were injected with vehicle formulated with various concentrations of phenol via a jugular vein cannula. Significant release of histamine was detected in blood samples taken 2min after dosing at the highest concentrations tested.


Asunto(s)
Degranulación de la Célula/efectos de los fármacos , Excipientes/toxicidad , Hematínicos/toxicidad , Histamina/metabolismo , Mastocitos/efectos de los fármacos , Péptidos/toxicidad , Fenol/toxicidad , Animales , Células Cultivadas , Química Farmacéutica , Relación Dosis-Respuesta a Droga , Excipientes/administración & dosificación , Excipientes/química , Femenino , Hematínicos/química , Histamina/sangre , Humanos , Inyecciones Intravenosas , Mastocitos/metabolismo , Ratones Endogámicos NOD , Péptidos/química , Fenol/administración & dosificación , Fenol/química , Cultivo Primario de Células , Ratas Sprague-Dawley , Medición de Riesgo , Factores de Tiempo
12.
J Virol ; 78(15): 8210-8, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254192

RESUMEN

Recombinant Listeria monocytogenes has many attractive characteristics as a vaccine vector against human immunodeficiency virus (HIV). Wild-type and attenuated Listeria strains expressing HIV Gag have been shown to induce long-lived mucosal and systemic T-cell responses in mice. Using the feline immunodeficiency virus (FIV) model of HIV we evaluated recombinant L. monocytogenes in a challenge system. Five cats were immunized with recombinant L. monocytogenes that expresses the FIV Gag and delivers an FIV Env-expressing DNA vaccine (LMgag/pND14-Lc-env). Control cats were either sham immunized or immunized with wild-type L. monocytogenes (LM-wt). At 1 year after vaginal challenge, provirus could not be detected in any of the nine tissues evaluated from cats immunized with the recombinant bacteria but was detected in at least one tissue in 8 of 10 control animals. Virus was isolated from bone marrow of four of five LMgag/pND14-Lc-env-immunized cats by use of a stringent coculture system but required CD8(+) T-cell depletion, indicating CD8(+) T-cell suppression of virus replication. Control animals had an inverted CD4:CD8 ratio in mesenteric lymph node and were depleted of both CD4(+) and CD8(+) intestinal epithelial T cells, while LMgag/pND14-Lc-env-immunized animals showed no such abnormalities. Vaginal FIV-specific immunoglobulin A was present at high titer in three LMgag/pND14-Lc-env-immunized cats before challenge and in all five at 1 year postchallenge. This study demonstrates that recombinant L. monocytogenes conferred some control of viral load after vaginal challenge with FIV.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida del Felino/prevención & control , Productos del Gen gag/inmunología , Virus de la Inmunodeficiencia Felina/aislamiento & purificación , Listeria monocytogenes/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología , Administración Oral , Animales , Anticuerpos Antivirales/biosíntesis , Gatos , Síndrome de Inmunodeficiencia Adquirida del Felino/virología , Femenino , Productos del Gen gag/genética , Inmunización , Inmunoglobulina A/biosíntesis , Inmunoglobulina G/biosíntesis , Mucosa Intestinal/inmunología , Ganglios Linfáticos/inmunología , Depleción Linfocítica , Provirus/aislamiento & purificación , Carga Viral
13.
Vet Immunol Immunopathol ; 95(3-4): 155-63, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12963276

RESUMEN

Striped bass (Morone saxatilis) and hybrid tilapia (Oreochromis spp.) were experimentally infected with Mycobacterium marinum. Splenic mononuclear cell transforming growth factor-beta (TGF-beta) mRNA was measured by reverse transcription quantitative-competitive PCR (RT-qcPCR). In histologic sections of liver and anterior kidney, the area of each section that was occupied by granulomas and the total area of each section were measured by computer-assisted image analysis and compared as a proportion (the granuloma proportion). Infected striped bass splenic mononuclear cell TGF-beta mRNA expression was significantly lower than uninfected controls, while for tilapia there was no significant difference between infected and control fish. Mycobacterial granuloma proportion of liver and anterior kidney sections was significantly greater for infected striped bass than tilapia. Three (of 10) infected tilapia with the most pronounced inflammatory response displayed a decrease in TGF-beta mRNA expression, similar to the overall striped bass response to mycobacterium challenge. Downregulation of TGF-beta and failure to modulate the immune response may be related to excessive inflammatory damage to organs observed in mycobacteria-sensitive fish species.


Asunto(s)
Lubina , Enfermedades de los Peces/inmunología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Infecciones por Mycobacterium no Tuberculosas/veterinaria , Mycobacterium marinum/inmunología , Tilapia , Factor de Crecimiento Transformador beta/inmunología , Animales , Enfermedades de los Peces/genética , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/patología , Regulación de la Expresión Génica/inmunología , Predisposición Genética a la Enfermedad , Granuloma/inmunología , Granuloma/microbiología , Granuloma/patología , Procesamiento de Imagen Asistido por Computador , Riñón/inmunología , Riñón/microbiología , Riñón/patología , Hígado/inmunología , Hígado/microbiología , Hígado/patología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Infecciones por Mycobacterium no Tuberculosas/patología , Mycobacterium marinum/genética , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA