Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cytotherapy ; 25(5): 490-501, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36781360

RESUMO

B-cell maturation antigen (BCMA) is a clinically validated target for multiple myeloma. T-cell engineered with chimeric antigen receptors (CARs) directed against BCMA have demonstrated robust therapeutic activity in clinical trials, but toxicities remain a significant concern for a subset of patients, supporting continued investigation of other engineered T-cell platforms that may offer equal efficacy with an improved toxicity profile. The authors recently described a BCMA-specific, T-cell-centric synthetic antigen receptor, the T-cell antigen coupler (TAC) receptor, that can be used to engineer T-cell with robust anti-myeloma activity. Here the authors describe the creation of a fully humanized BCMA-specific TAC receptor. Single-chain variable fragments (scFvs) were developed from BCMA-specific F(ab)s that were identified in a fully human phage display library. Twenty-four configurations of the F(ab)s were evaluated in a medium-throughput screening using primary T-cell, and a single F(ab), TRAC 3625, emerged as the most robust following in vitro and in vivo evaluation. An optimized BCMA-specific TAC receptor was developed through iterations of the BCMA-TAC design that evaluated a next-generation TAC scaffold sequence, different domains connecting the TAC to the 3625 scFv and different orientations of the TRAC 3625 heavy and light variable regions.


Assuntos
Mieloma Múltiplo , Linfócitos T , Humanos , Mieloma Múltiplo/terapia , Antígeno de Maturação de Linfócitos B , Imunoterapia Adotiva , Receptores de Antígenos de Linfócitos T
2.
Mol Carcinog ; 52(2): 103-17, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22086874

RESUMO

Telomerase is a ribonucleoprotein that maintains the ends of chromosomes (telomeres). In normal cells lacking telomerase activity, telomeres shorten with each cell division because of the inability to completely synthesize the lagging strand. Critically shortened telomeres elicit DNA damage responses and limit cellular division and lifespan, providing an important tumor suppressor function. Most human cancer cells express telomerase which contributes significantly to the tumor phenotype. In human breast cancer, telomerase expression is predictive of clinical outcomes such as lymph node metastasis and survival. In mouse models of mammary cancer, telomerase expression is also upregulated. Telomerase overexpression resulted in spontaneous mammary tumor development in aged female mice. Increased mammary cancer also was observed when telomerase deficient mice were crossed with p53 null mutant animals. However, the effects of telomerase and telomere length on oncogene driven mammary cancer have not been completely characterized. To address these issues we characterized neu proto-oncogene driven mammary tumor formation in G1 Terc-/- (telomerase deficient with long telomeres), G3 Terc-/- (telomerase deficient with short telomeres), and Terc+/+ mice. Telomerase deficiency reduced the number of mammary tumors and increased tumor latency regardless of telomere length. Decreased tumor formation correlated with increased apoptosis in Terc deficient tumors. Short telomeres dramatically increased lung metastasis which correlated with increased genomic instability, and specific alterations in DNA copy number and gene expression. We concluded that short telomeres promote metastasis in the absence of telomerase activity in neu oncogene driven mammary tumors.


Assuntos
Genes erbB-2 , Instabilidade Genômica , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Telomerase/metabolismo , Telômero , Animais , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proto-Oncogene Mas , RNA/genética , RNA/metabolismo , Telomerase/genética
3.
Oncoimmunology ; 10(1): 1995168, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777917

RESUMO

Engineered T cell therapies have revolutionized modern oncology, however processes for manufacturing T cell therapies vary and the impact of manufacturing processes On the cell product is poorly understood. Herein, we have used a commercially available hollow fiber membrane bioreactor (HFMBR) operated in a novel mode to demonstrate that T cells can be engineered with lentiviruses, grown to very high densities, and washed and harvested in a single, small volume bioreactor that is readily amenable to automation. Manufacturing within the HFMBR dramatically changed the programming of the T cells and yielded a product with greater therapeutic potency than T cells produced using the standard manual method. This change in programming was associated with increased resistance to cryopreservation, which is beneficial as T cell products are typically cryopreserved prior to administration to the patient. Transcriptional profiling of the T cells revealed a shift toward a glycolytic metabolism, which may protect cells from oxidative stress offering an explanation for the improved resistance to cryopreservation. This study reveals that the choice of bioreactor fundamentally impacts the engineered T cell product and must be carefully considered. Furthermore, these data challenge the premise that glycolytic metabolism is detrimental to T cell therapies.


Assuntos
Reatores Biológicos , Linfócitos T , Humanos
4.
Nat Commun ; 9(1): 3049, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-30076299

RESUMO

Engineering T cells with chimeric antigen receptors (CARs) is an effective method for directing T cells to attack tumors, but may cause adverse side effects such as the potentially lethal cytokine release syndrome. Here the authors show that the T cell antigen coupler (TAC), a chimeric receptor that co-opts the endogenous TCR, induces more efficient anti-tumor responses and reduced toxicity when compared with past-generation CARs. TAC-engineered T cells induce robust and antigen-specific cytokine production and cytotoxicity in vitro, and strong anti-tumor activity in a variety of xenograft models including solid and liquid tumors. In a solid tumor model, TAC-T cells outperform CD28-based CAR-T cells with increased anti-tumor efficacy, reduced toxicity, and faster tumor infiltration. Intratumoral TAC-T cells are enriched for Ki-67+ CD8+ T cells, demonstrating local expansion. These results indicate that TAC-T cells may have a superior therapeutic index relative to CAR-T cells.


Assuntos
Receptores de Antígenos/imunologia , Receptores de Antígenos Quiméricos/imunologia , Proteínas Recombinantes/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Antígenos CD28/imunologia , Linhagem Celular Tumoral , Citocinas/sangue , Citocinas/metabolismo , Citotoxicidade Imunológica , Feminino , Engenharia Genética , Células HEK293 , Humanos , Imunoterapia Adotiva/métodos , Lentivirus/genética , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos NOD , Engenharia de Proteínas , Receptor ErbB-2/imunologia , Receptores de Antígenos/genética , Receptores de Antígenos Quiméricos/genética , Anticorpos de Domínio Único , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Linfócitos T Citotóxicos/imunologia , Visão Ocular , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncotarget ; 8(19): 32101-32116, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28404880

RESUMO

Breast tumors comprise an infrequent tumor cell population, termed breast tumor initiating cells (BTIC), which sustain tumor growth, seed metastases and resist cytotoxic therapies. Hence therapies are needed to target BTIC to provide more durable breast cancer remissions than are currently achieved. We previously reported that serotonergic system antagonists abrogated the activity of mouse BTIC resident in the mammary tumors of a HER2-overexpressing model of breast cancer. Here we report that antagonists of serotonin (5-hydroxytryptamine; 5-HT) biosynthesis and activity, including US Federal Food and Drug Administration (FDA)-approved antidepressants, targeted BTIC resident in numerous breast tumor cell lines regardless of their clinical or molecular subtype. Notably, inhibitors of tryptophan hydroxylase 1 (TPH1), required for 5-HT biosynthesis in select non-neuronal cells, the serotonin reuptake transporter (SERT) and several 5-HT receptors compromised BTIC activity as assessed by functional sphere-forming assays. Consistent with these findings, human breast tumor cells express TPH1, 5-HT and SERT independent of their molecular or clinical subtype. Exposure of breast tumor cells ex vivo to sertraline (Zoloft), a selective serotonin reuptake inhibitor (SSRI), reduced BTIC frequency as determined by transplanting drug-treated tumor cells into immune-compromised mice. Moreover, another SSRI (vilazodone; Viibryd) synergized with chemotherapy to shrink breast tumor xenografts in immune-compromised mice by inhibiting tumor cell proliferation and inducing their apoptosis. Collectively our data suggest that antidepressants in combination with cytotoxic anticancer therapies may be an appropriate treatment regimen for testing in clinical trials.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Antagonistas da Serotonina/farmacologia , Serotonina/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Variações do Número de Cópias de DNA , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Expressão Gênica , Humanos , Camundongos , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triptofano Hidroxilase/genética , Triptofano Hidroxilase/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Oncol ; 38(4): 1175-81, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21305252

RESUMO

Telomeres are nucleoprotein structures at the ends of chromosomes that are composed of a repetitive G rich sequence and telomeric binding proteins. Telomeres prevent the degradation of chromosomal ends and protect against inappropriate recombination. Telomere attrition involves a tumor suppressor pathway that limits the replication of premalignant cells. The loss of telomeric DNA with each round of replication leads to growth arrest accompanied by senescence or apoptosis. Many tumor cells activate the telomerase gene to bypass senescence. Telomerase is a multisubunit ribonucleoprotein that uses an RNA template to catalyze the addition of telomeric DNA to chromosomal ends. Overexpression of the TERT subunit leads to telomere lengthening and extension of the replicative lifespan. Dominant-negative telomerase has been shown to inhibit telomerase activity in many tumor cell lines, and this is associated with telomere shortening and apoptosis. Additionally, pharmacological telomerase inhibitors have been developed which lead to progressive telomere shortening and programmed cell death. In this study, we report a series of human squamous cell carcinoma cell lines that have high telomerase activity and short telomeres. Dominant-negative telomerase expression and pharmacological telomerase inhibition failed to completely inhibit enzymatic activity which was accompanied by the lack of telomere shortening. These cells continued to proliferate and demonstrated fewer responsive genes when treated with a pharmacological telomerase inhibitor. We concluded that some human squamous cell carcinoma cell lines are resistant to telomerase inhibition.


Assuntos
Aminobenzoatos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteínas Mutantes/biossíntese , Naftalenos/farmacologia , Proteínas Recombinantes/biossíntese , Telomerase/antagonistas & inibidores , Apoptose , Carcinoma de Células Escamosas , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes , Genes Dominantes , Humanos , Fenótipo , Telomerase/biossíntese , Telomerase/genética
7.
Mol Cancer Res ; 9(7): 901-13, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21593138

RESUMO

Squamous cell carcinoma arises from highly proliferative basal layer epithelial cells, which normally divide for a short time before detaching from the basement membrane and undergoing terminal differentiation. Basal layer cells in stratified epithelia express the reverse transcriptase known as telomerase. Most human cells do not express telomerase and therefore are subject to loss of telomeric DNA with age due to the inability of lagging strand synthesis to completely replicate chromosomal ends. Late generation telomerase deficient mice exhibit signs of premature aging including reduced function of proliferating cellular compartments. We examined development of squamous cell carcinoma in a telomerase deficient murine background with long and short telomeres. G1 Terc-/- mice (long telomeres) had fewer lymph node metastases, which correlated with increased numbers of apoptotic cells in these tumors compared with wild-type mice. However, G5 Terc-/- mice with short telomeres had increased metastatic tumor burden similar to wild type mice. This increased metastasis correlated with genomic instability and aneuploidy in tumor cells from G5 Terc-/- mice. A number of similarities with human SCC were noted in the mouse model, and dramatic differences in global gene expression profiles were shown between primary and metastatic tumors. We concluded that telomere shortening promotes metastatic tumor development in a Terc null mouse model of head and neck cancer.


Assuntos
Carcinoma de Células Escamosas/secundário , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias Experimentais/patologia , Telomerase/genética , Telômero/genética , Animais , Carcinoma de Células Escamosas/genética , Perfilação da Expressão Gênica , Instabilidade Genômica , Neoplasias de Cabeça e Pescoço/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neoplasias Experimentais/genética
8.
Int J Oncol ; 36(3): 715-23, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20126992

RESUMO

The incidence of malignant tumors increases with age. This may be due to the duration of carcinogenesis or age related changes providing a favorable environment for tumor formation. Aging is associated with molecular, cellular and physiological events that influence carcinogenesis and cancer growth. Physiologic cell proliferation, differentiation, and aging can result in cell death. However, under the influence of exogenous or endogenous factors cells can undergo pathologic dedifferentiation, immortalization, and neoplastic clone formation. The effects of age have been recognized in both animal and human malignancies. These processes can result in cellular senescence as a barrier to tumorigenesis. Inducing senescence is an important outcome for the successful treatment of cancers particularly those resistant to apoptosis. Senescence is associated with polyploidy in several human cell lines. Polyploid cells are dangerous in that they can undergo aberrant mitoses giving rise to unstable progeny. Polyploid cells have been shown to escape senescence and divide. We examined the effects of aging on squamous cell carcinoma formation in a mouse model. Chronologically aged mice experience shorter tumor latency periods than wild-type animals. Tumors in aged mice were poorly vascularized, necrotic, and produced significantly fewer cervical lymph node metastases. Vascular endothelial growth factor expression was similar in primary tumors from young and old mice, but microvessel density was significantly reduced in tumors arising in aged mice. These results indicate that host response to angiogenic factors inhibit tumor growth and metastasis of head and neck cancer.


Assuntos
Envelhecimento , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neovascularização Patológica , Animais , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Poliploidia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa