Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
MAbs ; 15(1): 2191301, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36998195

RESUMO

There is considerable interest in the pharmaceutical industry toward development of antibody-based biotherapeutics because they can selectively bind diverse receptors and often possess desirable pharmacology. Here, we studied product characteristics of 89 marketed antibody-based biotherapeutics that were approved from 1986 to mid-2020 by gathering publicly available information. Our analyses revealed major trends in their emergence as the best-selling class of pharmaceuticals. Early on, most therapeutic monoclonal antibodies were developed to treat cancer, with CD20 being the most common target. Thanks to industrialization of antibody manufacturing technologies, their use has now blossomed to include 15 different therapeutic areas and nearly 60 targets, and the field is still growing! Drug manufacturers are solidifying their choices regarding types of antibodies and their molecular formats. IgG1 kappa continues to be the most common molecular format among marketed antibody-based biotherapeutics. Most antibody-based biotherapeutics approved since 2015 are either humanized or fully human, but the data we collected do not show a direct correlation between humanness and reported incidence of anti-drug antibodies. Furthermore, there have also been improvements in terms of drug product stability and high concentration liquid formulations suitable for subcutaneous route of administration, which are being approved more often in recent years. These improvements, however, have not been uniformly adopted across all therapeutic areas, suggesting that multiple options for drug product development are being used to serve diverse therapeutic purposes. Insights gained from this analysis may help us devise better end-to-end antibody-based biotherapeutic drug discovery and development strategies.


Assuntos
Desenvolvimento Industrial , Neoplasias , Humanos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/farmacologia , Neoplasias/tratamento farmacológico , Injeções Subcutâneas
2.
Mol Ther Methods Clin Dev ; 28: 190-207, 2023 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-36700123

RESUMO

Viral replication places oncolytic viruses (OVs) in a unique niche in the field of drug pharmacokinetics (PK) as their self-amplification obscures exposure-response relationships. Moreover, standard bioanalytical techniques are unable to distinguish the input from replicated drug products. Here, we combine two novel approaches to characterize PK and biodistribution (BD) after systemic administration of vesicular stomatitis virus pseudotyped with lymphocytic choriomeningitis virus glycoprotein (VSV-GP) in healthy mice. First: to decouple input drug PK/BD versus replication PK/BD, we developed and fully characterized a replication-incompetent tool virus that retained all other critical attributes of the drug. We used this approach to quantify replication in blood and tissues and to determine its impact on PK and BD. Second: to discriminate the genomic and antigenomic viral RNA strands contributing to replication dynamics in tissues, we developed an in situ hybridization method using strand-specific probes and assessed their spatiotemporal distribution in tissues. This latter approach demonstrated that distribution, transcription, and replication localized to tissue-resident macrophages, indicating their role in PK and BD. Ultimately, our study results in a refined PK/BD profile for a replicating OV, new proposed PK parameters, and deeper understanding of OV PK/BD using unique approaches that could be applied to other replicating vectors.

3.
Drug Discov Today ; 28(2): 103440, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36375739

RESUMO

Harnessing the immune system to kill tumors has been revolutionary and, as a result, has had an enormous benefit for patients in extending life and resulting in effective cures in some. However, activation of the immune system can come at the cost of undesirable adverse events such as cytokine release syndrome, immune-related adverse events, on-target/off-tumor toxicity, neurotoxicity and tumor lysis syndrome, which are safety risks that can be challenging to assess non-clinically. This article provides a review of the biology and mechanisms that can result in immune-mediated adverse effects and describes industry approaches using in vitro and in vivo models to aid in the nonclinical safety risk assessments for immune-oncology modalities. Challenges and limitations of knowledge and models are also discussed.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Medição de Risco
4.
Int J Pharm ; 609: 121162, 2021 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-34624444

RESUMO

Antibodies targeting the CD40-CD40L pathway have great potential for treating autoimmune diseases like rheumatoid arthritis, systemic lupus erythematosus (SLE), lupus nephritis (LN), and inflammatory bowel diseases (IBD). However, in addition to the known difficulty in generating a purely antagonistic CD40 antibody, the presence of CD40 and CD40L on platelets creates additional unique challenges for the safety, target coverage, and clearance of antibodies targeting this pathway. Previously described therapeutic antibodies targeting this pathway have various shortcomings, and the full therapeutic potential of this axis has yet to be realized. Herein, we describe the generation and characterization of BI 655064, a novel, purely antagonistic anti-CD40 antibody that potently neutralizes CD40-CD40L-dependent B-cell stimulation without evidence of impacting platelet functions. This uniquely optimized antibody targeting a highly challenging pathway was obtained by applying stringent functional and biophysical criteria during the lead selection process. BI 655064 has favorable target-mediated drug disposition (TMDD)-saturation pharmacokinetics, consistent with that of a high-quality therapeutic monoclonal antibody.


Assuntos
Doenças Autoimunes , Lúpus Eritematoso Sistêmico , Doenças Autoimunes/tratamento farmacológico , Linfócitos B , Antígenos CD40 , Ligante de CD40 , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico
5.
Regul Toxicol Pharmacol ; 98: 98-107, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30026135

RESUMO

Nonclinical safety testing of biopharmaceuticals can present significant challenges to human risk assessment with these innovative and often complex drugs. Emerging topics in this field were discussed recently at the 2016 Annual US BioSafe General Membership meeting. The presentations and subsequent discussions from the main sessions are summarized. The topics covered included: (i) specialty biologics (oncolytic virus, gene therapy, and gene editing-based technologies), (ii) the value of non-human primates (NHPs) for safety assessment, (iii) challenges in the safety assessment of immuno-oncology drugs (T cell-dependent bispecifics, checkpoint inhibitors, and costimulatory agonists), (iv) emerging therapeutic approaches and modalities focused on microbiome, oligonucleotide, messenger ribonucleic acid (mRNA) therapeutics, (v) first in human (FIH) dose selection and the minimum anticipated biological effect level (MABEL), (vi) an update on current regulatory guidelines, International Council for Harmonization (ICH) S1, S3a, S5, S9 and S11 and (vii) breakout sessions that focused on bioanalytical and PK/PD challenges with bispecific antibodies, cytokine release in nonclinical studies, determining adversity and NOAEL for biologics, the value of second species for toxicology assessment and what to do if there is no relevant toxicology species.


Assuntos
Produtos Biológicos/toxicidade , Avaliação Pré-Clínica de Medicamentos/métodos , Animais , Anticorpos Monoclonais/toxicidade , Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Humanos , Proteínas Recombinantes/toxicidade , Medição de Risco
6.
PLoS One ; 9(8): e105883, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25170619

RESUMO

Inflammation is associated with immune cells infiltrating into the inflammatory site and pain. CC chemokine receptor 1 (CCR1) mediates trafficking of leukocytes to sites of inflammation. However, the contribution of CCR1 to pain is incompletely understood. Here we report an unexpected discovery that CCR1-mediated trafficking of neutrophils and CCR1 activity on non-hematopoietic cells both modulate pain. Using a genetic approach (CCR1-/- animals) and pharmacological inhibition of CCR1 with selective inhibitors, we show significant reductions in pain responses using the acetic acid-induced writhing and complete Freund's adjuvant-induced mechanical hyperalgesia models. Reductions in writhing correlated with reduced trafficking of myeloid cells into the peritoneal cavity. We show that CCR1 is highly expressed on circulating neutrophils and their depletion decreases acetic acid-induced writhing. However, administration of neutrophils into the peritoneal cavity did not enhance acetic acid-induced writhing in wild-type (WT) or CCR1-/- mice. Additionally, selective knockout of CCR1 in either the hematopoietic or non-hematopoietic compartments also reduced writhing. Together these data suggest that CCR1 functions to significantly modulate pain by controlling neutrophil trafficking to the inflammatory site and having an unexpected role on non-hematopoietic cells. As inflammatory diseases are often accompanied with infiltrating immune cells at the inflammatory site and pain, CCR1 antagonism may provide a dual benefit by restricting leukocyte trafficking and reducing pain.


Assuntos
Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Dor/imunologia , Receptores CCR1/imunologia , Ácido Acético , Animais , Artrite Experimental/genética , Artrite Experimental/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Movimento Celular/genética , Movimento Celular/imunologia , Citometria de Fluxo , Adjuvante de Freund , Hiperalgesia/induzido quimicamente , Hiperalgesia/genética , Hiperalgesia/imunologia , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Infiltração de Neutrófilos/genética , Neutrófilos/metabolismo , Dor/induzido quimicamente , Dor/genética , Medição da Dor/métodos , Peritonite/genética , Peritonite/imunologia , Peritonite/metabolismo , Receptores CCR1/antagonistas & inibidores , Receptores CCR1/genética
7.
Mol Med ; 17(3-4): 211-20, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21107497

RESUMO

It is clear that estrogen can accelerate and exacerbate disease in some lupus-prone mouse strains. It also appears that estrogen can contribute to disease onset or flare in a subset of patients with lupus. We have previously shown estrogen alters B-cell development to decrease lymphopoiesis and increase the frequency of marginal zone B cells. Furthermore, estrogen diminishes B-cell receptor signaling and allows for the increased survival of high-affinity DNA-reactive B cells. Here, we analyze the contribution of estrogen receptor α or ß engagement to the altered B-cell maturation and selection mediated by increased exposure to estrogen. We demonstrate that engagement of either estrogen receptor α or ß can alter B-cell maturation, but only engagement of estrogen receptor α is a trigger for autoimmunity. Thus, maturation and selection are regulated differentially by estrogen. These observations have therapeutic implications.


Assuntos
Linfócitos B/imunologia , Receptor alfa de Estrogênio/imunologia , Receptor beta de Estrogênio/imunologia , Transdução de Sinais/imunologia , Animais , Fator Ativador de Células B/genética , Fator Ativador de Células B/imunologia , Fator Ativador de Células B/metabolismo , Subpopulações de Linfócitos B/efeitos dos fármacos , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Western Blotting , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Estrogênios/farmacologia , Feminino , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovariectomia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
J Autoimmun ; 32(3-4): 149-57, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19327966

RESUMO

FcgammaRIIB is an inhibitory receptor which plays a role in limiting B cell and DC activation. Since FcgammaRIIB is known to dampen the signaling strength of the BCR, we wished to determine the impact of FcgammaRIIB on the regulation of BCRs which differ in their affinity for DNA. For these studies, FcgammaRIIB deficient BALB/c mice were bred with mice expressing the transgene-encoded H chain of the R4A anti-DNA antibody which gives rise to BCRs which express high, low or no affinity for DNA. The deletion of FcgammaRIIB in R4A BALB/c mice led to an alteration in the B cell repertoire, allowing for the expansion and activation of high affinity DNA-reactive B cells. By 6-8 months of age, R4A x FcgammaRIIB-/- BALB/c mice spontaneously developed anti-DNA antibody titers. These mice also displayed an induction of IFN-inducible genes and an elevation in levels of the B cell survival factor, BAFF. These data demonstrate that FcgammaRIIB preferentially limits activation of high affinity autoreactive B cells and can influence the activation of DC through an immune complex-mediated mechanism.


Assuntos
Anticorpos Antinucleares/imunologia , Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Células Dendríticas/imunologia , Receptores de IgG/metabolismo , Animais , Anticorpos Antinucleares/genética , Anticorpos Antinucleares/metabolismo , Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Interferons/genética , Interferons/imunologia , Interferons/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de IgG/genética , Receptores de IgG/imunologia
10.
Curr Opin Rheumatol ; 18(5): 456-61, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16896282

RESUMO

PURPOSE OF REVIEW: For many decades, it has been speculated that sex hormones play a role in systemic lupus erythematosus. Recent data accumulated during the past few years provide striking evidence that hormonal modulation of B cells can have a profound impact on the survival, maturation and repertoire selection of autoreactive B cells and begin to explain the sex bias associated with the condition. RECENT FINDINGS: While there are still insufficient clinical data to define a role for estrogen or prolactin in human systemic lupus erythematosus, recent studies of anti-DNA antibody transgenic mice clearly demonstrate that an elevation in either estrogen or prolactin breaks tolerance of high affinity DNA-reactive B cells and induces a lupus phenotype. B cells with the same antigenic specificities are rescued by either estrogen or prolactin, but estrogen promotes the survival and activation of the T independent marginal zone B cell subset, while prolactin promotes the survival and activation of the T dependent follicular B cell subset. SUMMARY: Elevations in the levels of estrogen or prolactin can promote the survival and activation of high affinity autoreactive B cells. These hormones engage different B cell pathways to interfere with B cell tolerance. The identification of systemic lupus erythematosus patients with either an estrogen-responsive or prolactin-responsive disease will further the development of therapeutics that can specifically modulate hormonal responses.


Assuntos
Estrogênios/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Prolactina/imunologia , Tolerância a Antígenos Próprios/imunologia , Caracteres Sexuais , Transdução de Sinais/imunologia , Animais , Anticorpos Antinucleares/genética , Anticorpos Antinucleares/imunologia , Linfócitos B/imunologia , Linfócitos B/patologia , Sobrevivência Celular/imunologia , Feminino , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Lúpus Eritematoso Sistêmico/terapia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Tolerância a Antígenos Próprios/genética , Transdução de Sinais/genética , Linfócitos T/imunologia , Linfócitos T/patologia
11.
J Immunol ; 176(5): 2703-10, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16493025

RESUMO

There are increasing data suggesting that sex hormones, such as estrogen, have immunomodulatory effects and play a role in disease progression and pathogenesis in patients with the autoimmune disorder systemic lupus erythematosus. We have shown previously that treatment with 17beta-estradiol (E2) induces a lupus phenotype in BALB/c mice that express a transgene-encoded H chain of an anti-DNA Ab. Because E2 treatment interferes with normal tolerance of naive DNA-reactive B cells, we elected to study the effects of hormonal modulation on the regulation of autoreactive B cells at early developmental checkpoints. Single-cell PCR was performed to study the repertoire of DNA-reactive B cell subsets. High-affinity DNA-reactive B cells were rescued at both the immature and transitional B cell stage in E2-treated mice. Interestingly, although low-affinity DNA-reactive B cells survive negative selection in control mice, the frequency of these cells was significantly reduced in the mature pool of E2-treated mice, suggesting that the high-affinity DNA-reactive cells that mature to immunocompetence out-compete the low-affinity population for survival as mature B cells. These data provide evidence that an elevation in serum levels of E2 facilitates the maturation of a pathogenic naive autoreactive B cell repertoire and hampers the maturation of a potentially protective autoreactive B cell repertoire. Furthermore, these data show that both positive and negative selection occur within the transitional B cell stage.


Assuntos
Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/metabolismo , Diferenciação Celular/imunologia , DNA/imunologia , Estradiol/farmacologia , Animais , Subpopulações de Linfócitos B/imunologia , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/imunologia , Antígenos CD4/genética , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , DNA/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia
12.
Mol Immunol ; 42(7): 811-20, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15829269

RESUMO

Systemic lupus erythematosus is an autoimmune disorder characterized by the production of pathogenic autoantibodies, primarily to nuclear antigens. The etiology of SLE is not entirely understood, but it is well-appreciated that multiple factors such as genetics and environment contribute to disease progression and pathogenesis. There is also convincing evidence that gender plays an import role in SLE since the incidence of disease occurs with a female to male ratio of 9:1. While it is plausible that some sex-linked genes may contribute to the genetic predisposition for the disease, other likely culprits for this gender bias are the sex hormones estrogen and prolactin. The data implicating estrogen and prolactin in SLE, until recently, were largely circumstantial. However, within the last few years, data collected from both human and mouse studies have provided compelling evidence that alterations in sex hormone levels can alter tolerance of autoreactive B cells and exacerbate disease. In this review, we will discuss recent data demonstrating a role for estrogen and prolactin in SLE and the effect of these hormones on B cell maturation, selection and activation.


Assuntos
Linfócitos B/imunologia , Estrogênios/imunologia , Lúpus Eritematoso Sistêmico/etiologia , Prolactina/imunologia , Animais , Linfócitos B/efeitos dos fármacos , Diferenciação Celular , Estrogênios/metabolismo , Estrogênios/farmacologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Prolactina/metabolismo , Prolactina/farmacologia
13.
J Immunol ; 173(5): 3524-34, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15322217

RESUMO

B cell-activating factor belonging to the TNF family (BAFF) blockade prevents the onset of disease in systemic lupus erythematosus (SLE)-prone NZB/NZW F(1) mice. To determine the mechanism of this effect, we administered a short course of TACI-Ig with and without six doses of CTLA4-Ig to 18- to 20-wk-old NZB/NZW F(1) mice and evaluated the effect on B and T cell subsets and on anti-dsDNA Ab-producing B cells. Even a brief exposure to TACI-Ig had a beneficial effect on murine SLE; CTLA4-Ig potentiated this effect. The combination of TACI-Ig and CTLA4-Ig resulted in a temporary decrease in serum IgG levels. However, after cessation of treatment, high titers of IgG anti-dsDNA Abs appeared in the serum and IgG Abs deposited in the kidneys. Despite the appearance of pathogenic autoantibodies, the onset of proteinuria was markedly delayed; this was associated with prolonged depletion of B cells past the T1 stage, a decrease in the size of the spleen and lymph nodes, and a decrease in the absolute number of activated and memory CD4(+) T cells. TACI-Ig treatment normalized serum levels of IgM that are markedly elevated in NZB/W F(1) mice; this appeared to be due to a prolonged effect on the ability of the splenic microenvironment to support short-lived IgM plasma cells. Finally, a short course of combination TACI-Ig and CTLA4-Ig prolonged life and even reversed proteinuria in aged NZB/W F(1) mice, suggesting that BAFF blockade may be an effective therapeutic strategy for active SLE.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Proteínas de Membrana/imunologia , Fator de Necrose Tumoral alfa/imunologia , Abatacepte , Animais , Autoimunidade/imunologia , Fator Ativador de Células B , Linfócitos B/imunologia , Linfócitos B/fisiologia , Modelos Animais de Doenças , Imunoconjugados/genética , Imunoconjugados/imunologia , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Proteínas de Membrana/genética , Camundongos , Fenótipo , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteína Transmembrana Ativadora e Interagente do CAML , Fator de Necrose Tumoral alfa/genética
14.
J Clin Invest ; 109(12): 1625-33, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12070310

RESUMO

Estrogen is thought to contribute to the increased frequency of autoimmune disorders occurring in females, but a molecular basis for its effects on autoimmunity remains to be elucidated. We have shown previously that estrogen leads to the survival and activation of autoreactive cells in the naive repertoire. To identify the molecular pathways involved in B cell tolerance, we sought to identify genes that are differentially regulated by estrogen in mouse B cells. Several genes involved in B cell activation and survival, including cd22, shp-1, bcl-2, and vcam-1, were upregulated by estrogen in B cells. We found that overexpression of CD22 and SHP-1 in B cells decreased B cell receptor signaling. Estrogen receptors alpha and beta are expressed on B cells and are functional, since they can directly upregulate expression of CD22, SHP-1, and Bcl-2. Estrogen treatment protected isolated primary B cells from B cell receptor-mediated apoptosis. These results suggest that estrogen induces a genetic program that alters survival and activation of B cells in a B cell-autonomous fashion and thus skews the naive immune system toward autoreactivity.


Assuntos
Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos B/genética , Apoptose , Linfócitos B/metabolismo , Moléculas de Adesão Celular , Estradiol/farmacologia , Lectinas , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Molécula 1 de Adesão de Célula Vascular/genética , Animais , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Células Cultivadas , Estradiol/administração & dosagem , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Receptores de Estrogênio/genética , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA