Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Transl Med ; 16(755): eadg7123, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38985855

RESUMEN

Two types of engineered T cells have been successfully used to treat patients with cancer, one with an antigen recognition domain derived from antibodies [chimeric antigen receptors (CARs)] and the other derived from T cell receptors (TCRs). CARs use high-affinity antigen-binding domains and costimulatory domains to induce T cell activation but can only react against target cells with relatively high amounts of antigen. TCRs have a much lower affinity for their antigens but can react against target cells displaying only a few antigen molecules. Here, we describe a new type of receptor, called a Co-STAR (for costimulatory synthetic TCR and antigen receptor), that combines aspects of both CARs and TCRs. In Co-STARs, the antigen-recognizing components of TCRs are replaced by high-affinity antibody fragments, and costimulation is provided by two modules that drive NF-κB signaling (MyD88 and CD40). Using a TCR-mimic antibody fragment that targets a recurrent p53 neoantigen presented in a common human leukocyte antigen (HLA) allele, we demonstrate that T cells equipped with Co-STARs can kill cancer cells bearing low densities of antigen better than T cells engineered with conventional CARs and patient-derived TCRs in vitro. In mouse models, we show that Co-STARs mediate more robust T cell expansion and more durable tumor regressions than TCRs similarly modified with MyD88 and CD40 costimulation. Our data suggest that Co-STARs may have utility for other peptide-HLA antigens in cancer and other targets where antigen density may limit the efficacy of engineered T cells.


Asunto(s)
Neoplasias , Receptores de Antígenos de Linfocitos T , Receptores Quiméricos de Antígenos , Humanos , Animales , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Línea Celular Tumoral , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Transducción de Señal
2.
Nature ; 628(8007): 416-423, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538786

RESUMEN

Antibody and chimeric antigen receptor (CAR) T cell-mediated targeted therapies have improved survival in patients with solid and haematologic malignancies1-9. Adults with T cell leukaemias and lymphomas, collectively called T cell cancers, have short survival10,11 and lack such targeted therapies. Thus, T cell cancers particularly warrant the development of CAR T cells and antibodies to improve patient outcomes. Preclinical studies showed that targeting T cell receptor ß-chain constant region 1 (TRBC1) can kill cancerous T cells while preserving sufficient healthy T cells to maintain immunity12, making TRBC1 an attractive target to treat T cell cancers. However, the first-in-human clinical trial of anti-TRBC1 CAR T cells reported a low response rate and unexplained loss of anti-TRBC1 CAR T cells13,14. Here we demonstrate that CAR T cells are lost due to killing by the patient's normal T cells, reducing their efficacy. To circumvent this issue, we developed an antibody-drug conjugate that could kill TRBC1+ cancer cells in vitro and cure human T cell cancers in mouse models. The anti-TRBC1 antibody-drug conjugate may provide an optimal format for TRBC1 targeting and produce superior responses in patients with T cell cancers.


Asunto(s)
Inmunoconjugados , Leucemia de Células T , Linfoma de Células T , Receptores de Antígenos de Linfocitos T alfa-beta , Linfocitos T , Animales , Femenino , Humanos , Ratones , Inmunoconjugados/inmunología , Inmunoconjugados/uso terapéutico , Inmunoterapia Adoptiva , Leucemia de Células T/tratamiento farmacológico , Leucemia de Células T/inmunología , Linfoma de Células T/tratamiento farmacológico , Linfoma de Células T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Nat Commun ; 14(1): 5063, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37604828

RESUMEN

Specificity remains a major challenge to current therapeutic strategies for cancer. Mutation associated neoantigens (MANAs) are products of genetic alterations, making them highly specific therapeutic targets. MANAs are HLA-presented (pHLA) peptides derived from intracellular mutant proteins that are otherwise inaccessible to antibody-based therapeutics. Here, we describe the cryo-EM structure of an antibody-MANA pHLA complex. Specifically, we determine a TCR mimic (TCRm) antibody bound to its MANA target, the KRASG12V peptide presented by HLA-A*03:01. Hydrophobic residues appear to account for the specificity of the mutant G12V residue. We also determine the structure of the wild-type G12 peptide bound to HLA-A*03:01, using X-ray crystallography. Based on these structures, we perform screens to validate the key residues required for peptide specificity. These experiments led us to a model for discrimination between the mutant and the wild-type peptides presented on HLA-A*03:01 based exclusively on hydrophobic interactions.


Asunto(s)
Anticuerpos , Proteínas Proto-Oncogénicas p21(ras) , Proteínas Proto-Oncogénicas p21(ras)/genética , Reconocimiento en Psicología , Interacciones Hidrofóbicas e Hidrofílicas , Antígenos HLA-A/genética
4.
Cancer Discov ; 13(10): 2166-2179, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37565753

RESUMEN

Cell-free DNA (cfDNA) concentrations from patients with cancer are often elevated compared with those of healthy controls, but the sources of this extra cfDNA have never been determined. To address this issue, we assessed cfDNA methylation patterns in 178 patients with cancers of the colon, pancreas, lung, or ovary and 64 patients without cancer. Eighty-three of these individuals had cfDNA concentrations much greater than those generally observed in healthy subjects. The major contributor of cfDNA in all samples was leukocytes, accounting for ∼76% of cfDNA, with neutrophils predominating. This was true regardless of whether the samples were derived from patients with cancer or the total plasma cfDNA concentration. High levels of cfDNA observed in patients with cancer did not come from either neoplastic cells or surrounding normal epithelial cells from the tumor's tissue of origin. These data suggest that cancers may have a systemic effect on cell turnover or DNA clearance. SIGNIFICANCE: The origin of excess cfDNA in patients with cancer is unknown. Using cfDNA methylation patterns, we determined that neither the tumor nor the surrounding normal tissue contributes this excess cfDNA-rather it comes from leukocytes. This finding suggests that cancers have a systemic impact on cell turnover or DNA clearance. See related commentary by Thierry and Pisareva, p. 2122. This article is featured in Selected Articles from This Issue, p. 2109.


Asunto(s)
Ácidos Nucleicos Libres de Células , Neoplasias Colorrectales , Neoplasias Ováricas , Humanos , Femenino , Ácidos Nucleicos Libres de Células/genética , Metilación de ADN , ADN de Neoplasias/genética , Páncreas/patología , Neoplasias Ováricas/genética , Pulmón/patología , Neoplasias Colorrectales/genética , Biomarcadores de Tumor/genética
5.
Nat Cancer ; 2(5): 487-497, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34676374

RESUMEN

Several current immunotherapy approaches target private neoantigens derived from mutations that are unique to individual patients' tumors. However, immunotherapeutic agents can also be developed against public neoantigens derived from recurrent mutations in cancer driver genes. The latter approaches target proteins that are indispensable for tumor growth, and each therapeutic agent can be applied to numerous patients. Here we review the opportunities and challenges involved in the identification of suitable public neoantigen targets and the development of therapeutic agents targeting them.


Asunto(s)
Antígenos de Neoplasias , Neoplasias , Antígenos de Neoplasias/genética , Humanos , Factores Inmunológicos/uso terapéutico , Inmunoterapia , Mutación , Neoplasias/terapia , Oncogenes
6.
Nat Commun ; 12(1): 5271, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34489470

RESUMEN

Chimeric antigen receptor (CAR) T cells have emerged as a promising class of therapeutic agents, generating remarkable responses in the clinic for a subset of human cancers. One major challenge precluding the wider implementation of CAR therapy is the paucity of tumor-specific antigens. Here, we describe the development of a CAR targeting the tumor-specific isocitrate dehydrogenase 2 (IDH2) with R140Q mutation presented on the cell surface in complex with a common human leukocyte antigen allele, HLA-B*07:02. Engineering of the hinge domain of the CAR, as well as crystal structure-guided optimization of the IDH2R140Q-HLA-B*07:02-targeting moiety, enhances the sensitivity and specificity of CARs to enable targeting of this HLA-restricted neoantigen. This approach thus holds promise for the development and optimization of immunotherapies specific to other cancer driver mutations that are difficult to target by conventional means.


Asunto(s)
Antígeno HLA-B7/química , Isocitrato Deshidrogenasa/metabolismo , Ingeniería de Proteínas/métodos , Receptores Quiméricos de Antígenos/química , Animales , Antígenos de Neoplasias/metabolismo , Células COS , Línea Celular , Chlorocebus aethiops , Epítopos , Antígeno HLA-B7/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Isocitrato Deshidrogenasa/química , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/inmunología , Mutación , Biblioteca de Péptidos , Conformación Proteica , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/fisiología
7.
Sci Immunol ; 6(57)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33649101

RESUMEN

Mutations in the RAS oncogenes occur in multiple cancers, and ways to target these mutations has been the subject of intense research for decades. Most of these efforts are focused on conventional small-molecule drugs rather than antibody-based therapies because the RAS proteins are intracellular. Peptides derived from recurrent RAS mutations, G12V and Q61H/L/R, are presented on cancer cells in the context of two common human leukocyte antigen (HLA) alleles, HLA-A3 and HLA-A1, respectively. Using phage display, we isolated single-chain variable fragments (scFvs) specific for each of these mutant peptide-HLA complexes. The scFvs did not recognize the peptides derived from the wild-type form of RAS proteins or other related peptides. We then sought to develop an immunotherapeutic agent that was capable of killing cells presenting very low levels of these RAS-derived peptide-HLA complexes. Among many variations of bispecific antibodies tested, one particular format, the single-chain diabody (scDb), exhibited superior reactivity to cells expressing low levels of neoantigens. We converted the scFvs to this scDb format and demonstrated that they were capable of inducing T cell activation and killing of target cancer cells expressing endogenous levels of the mutant RAS proteins and cognate HLA alleles. CRISPR-mediated alterations of the HLA and RAS genes provided strong genetic evidence for the specificity of the scDbs. Thus, this approach could be applied to other common oncogenic mutations that are difficult to target by conventional means, allowing for more specific anticancer therapeutics.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antígenos de Neoplasias , Biomarcadores de Tumor/antagonistas & inhibidores , Proteínas Mutantes/antagonistas & inhibidores , Proteínas ras/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Anticuerpos Biespecíficos/inmunología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/química , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Línea Celular , Reacciones Cruzadas , Antígenos HLA/inmunología , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Proteínas Mutantes/química , Proteínas Mutantes/inmunología , Mutación , Fragmentos de Péptidos , Unión Proteica/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Proteínas ras/química , Proteínas ras/genética , Proteínas ras/inmunología
8.
Science ; 371(6533)2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33649166

RESUMEN

TP53 (tumor protein p53) is the most commonly mutated cancer driver gene, but drugs that target mutant tumor suppressor genes, such as TP53, are not yet available. Here, we describe the identification of an antibody highly specific to the most common TP53 mutation (R175H, in which arginine at position 175 is replaced with histidine) in complex with a common human leukocyte antigen-A (HLA-A) allele on the cell surface. We describe the structural basis of this specificity and its conversion into an immunotherapeutic agent: a bispecific single-chain diabody. Despite the extremely low p53 peptide-HLA complex density on the cancer cell surface, the bispecific antibody effectively activated T cells to lyse cancer cells that presented the neoantigen in vitro and in mice. This approach could in theory be used to target cancers containing mutations that are difficult to target in conventional ways.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Antineoplásicos/inmunología , Antígenos de Neoplasias/inmunología , Antígeno HLA-A2/inmunología , Neoplasias/terapia , Proteína p53 Supresora de Tumor/inmunología , Alelos , Animales , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Antineoplásicos/química , Anticuerpos Antineoplásicos/uso terapéutico , Arginina/genética , Células COS , Chlorocebus aethiops , Femenino , Células HEK293 , Antígeno HLA-A2/química , Antígeno HLA-A2/genética , Histidina/genética , Humanos , Inmunización Pasiva , Células Jurkat , Activación de Linfocitos , Ratones Endogámicos NOD , Mutación , Linfocitos T/inmunología , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Sci Transl Med ; 13(584)2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33649188

RESUMEN

Immunotherapies such as chimeric antigen receptor (CAR) T cells and bispecific antibodies redirect healthy T cells to kill cancer cells expressing the target antigen. The pan-B cell antigen-targeting immunotherapies have been remarkably successful in treating B cell malignancies. Such therapies also result in the near-complete loss of healthy B cells, but this depletion is well tolerated by patients. Although analogous targeting of pan-T cell markers could, in theory, help control T cell cancers, the concomitant healthy T cell depletion would result in severe and unacceptable immunosuppression. Thus, therapies directed against T cell cancers require more selective targeting. Here, we describe an approach to target T cell cancers through T cell receptor (TCR) antigens. Each T cell, normal or malignant, expresses a unique TCR ß chain generated from 1 of 30 TCR ß chain variable gene families (TRBV1 to TRBV30). We hypothesized that bispecific antibodies targeting a single TRBV family member expressed in malignant T cells could promote killing of these cancer cells, while preserving healthy T cells that express any of the other 29 possible TRBV family members. We addressed this hypothesis by demonstrating that bispecific antibodies targeting TRBV5-5 (α-V5) or TRBV12 (α-V12) specifically lyse relevant malignant T cell lines and patient-derived T cell leukemias in vitro. Treatment with these antibodies also resulted in major tumor regressions in mouse models of human T cell cancers. This approach provides an off-the-shelf, T cell cancer selective targeting approach that preserves enough healthy T cells to maintain cellular immunity.


Asunto(s)
Anticuerpos Biespecíficos , Trastornos Linfoproliferativos/terapia , Linfocitos T/patología , Humanos , Receptores de Antígenos de Linfocitos T alfa-beta
10.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33731480

RESUMEN

Developing therapeutic agents with potent antitumor activity that spare normal tissues remains a significant challenge. Clonal loss of heterozygosity (LOH) is a widespread and irreversible genetic alteration that is exquisitely specific to cancer cells. We hypothesized that LOH events can be therapeutically targeted by "inverting" the loss of an allele in cancer cells into an activating signal. Here we describe a proof-of-concept approach utilizing engineered T cells approximating NOT-gate Boolean logic to target counterexpressed antigens resulting from LOH events in cancer. The NOT gate comprises a chimeric antigen receptor (CAR) targeting the allele of human leukocyte antigen (HLA) that is retained in the cancer cells and an inhibitory CAR (iCAR) targeting the HLA allele that is lost in the cancer cells. We demonstrate that engineered T cells incorporating such NOT-gate logic can be activated in a genetically predictable manner in vitro and in mice to kill relevant cancer cells. This therapeutic approach, termed NASCAR (Neoplasm-targeting Allele-Sensing CAR), could, in theory, be extended to LOH of other polymorphic genes that result in altered cell surface antigens in cancers.


Asunto(s)
Biomarcadores de Tumor , Inmunoterapia , Pérdida de Heterocigocidad , Terapia Molecular Dirigida , Neoplasias/etiología , Neoplasias/terapia , Alelos , Antígenos de Neoplasias/inmunología , Tratamiento Basado en Trasplante de Células y Tejidos , Antígenos HLA/genética , Antígenos HLA/inmunología , Humanos , Inmunoterapia/métodos , Inmunoterapia Adoptiva , Terapia Molecular Dirigida/efectos adversos , Terapia Molecular Dirigida/métodos , Anticuerpos de Cadena Única/farmacología , Anticuerpos de Cadena Única/uso terapéutico
12.
PLoS One ; 15(12): e0233662, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33270630

RESUMEN

BACKGROUND: The effects of diet in cancer, in general, and breast cancer in particular, are not well understood. Insulin inhibition in ketogenic, high fat diets, modulate downstream signaling molecules and are postulated to have therapeutic benefits. Obesity and diabetes have been associated with higher incidence of breast cancer. Addition of anti-cancer drugs together with diet is also not well studied. METHODS: Two diets, one ketogenic, the other standard mouse chow, were tested in a spontaneous breast cancer model in 34 mice. Subgroups of 3-9 mice were assigned, in which the diet were implemented either with or without added rapamycin, an mTOR inhibitor and potential anti-cancer drug. RESULTS: Blood glucose and insulin concentrations in mice ingesting the ketogenic diet (KD) were significantly lower, whereas beta hydroxybutyrate (BHB) levels were significantly higher, respectively, than in mice on the standard diet (SD). Growth of primary breast tumors and lung metastases were inhibited, and lifespans were longer in the KD mice compared to mice on the SD (p<0.005). Rapamycin improved survival in both mouse diet groups, but when combined with the KD was more effective than when combined with the SD. CONCLUSIONS: The study provides proof of principle that a ketogenic diet a) results in serum insulin reduction and ketosis in a spontaneous breast cancer mouse model; b) can serve as a therapeutic anti-cancer agent; and c) can enhance the effects of rapamycin, an anti-cancer drug, permitting dose reduction for comparable effect. Further, the ketogenic diet in this model produces superior cancer control than standard mouse chow whether with or without added rapamycin.


Asunto(s)
Neoplasias de la Mama/dietoterapia , Neoplasias de la Mama/tratamiento farmacológico , Dieta Cetogénica/métodos , Sirolimus/farmacología , Ácido 3-Hidroxibutírico/metabolismo , Animales , Antineoplásicos/farmacología , Glucemia/efectos de los fármacos , Neoplasias de la Mama/sangre , Neoplasias de la Mama/metabolismo , Modelos Animales de Enfermedad , Femenino , Insulina/sangre , Cetosis/fisiopatología , Ratones
13.
Expert Rev Clin Immunol ; 16(12): 1185-1204, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33146561

RESUMEN

Introduction: The mortality of coronavirus disease 2019 (COVID-19) is frequently driven by an injurious immune response characterized by the development of acute respiratory distress syndrome (ARDS), endotheliitis, coagulopathy, and multi-organ failure. This spectrum of hyperinflammation in COVID-19 is commonly referred to as cytokine storm syndrome (CSS). Areas covered: Medline and Google Scholar were searched up until 15th of August 2020 for relevant literature. Evidence supports a role of dysregulated immune responses in the immunopathogenesis of severe COVID-19. CSS associated with SARS-CoV-2 shows similarities to the exuberant cytokine production in some patients with viral infection (e.g.SARS-CoV-1) and may be confused with other syndromes of hyperinflammation like the cytokine release syndrome (CRS) in CAR-T cell therapy. Interleukin (IL)-6, IL-8, and tumor necrosis factor-alpha have emerged as predictors of COVID-19 severity and in-hospital mortality. Expert opinion: Despite similarities, COVID-19-CSS appears to be distinct from HLH, MAS, and CRS, and the application of HLH diagnostic scores and criteria to COVID-19 is not supported by emerging data. While immunosuppressive therapy with glucocorticoids has shown a mortality benefit, cytokine inhibitors may hold promise as 'rescue therapies' in severe COVID-19. Given the arguably limited benefit in advanced disease, strategies to prevent the development of COVID-19-CSS are needed.


Asunto(s)
COVID-19 , Síndrome de Liberación de Citoquinas , Citocinas/sangre , SARS-CoV-2/metabolismo , Terapias en Investigación , COVID-19/sangre , COVID-19/mortalidad , COVID-19/prevención & control , COVID-19/terapia , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/mortalidad , Síndrome de Liberación de Citoquinas/prevención & control , Síndrome de Liberación de Citoquinas/terapia , Humanos
14.
PLoS One ; 14(2): e0211737, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30763339

RESUMEN

Triple negative breast cancer (TNBC) is an aggressive tumor with propensity to metastasize and poor treatment options. Improving treatment options would be impactful; thus, finding a tumor-specific cell surface protein with metastasis promoting functions that could be knocked out was the goal of this study. The Otoconin 90 gene (OC90), frequently amplified in tumors on chromosome 8q24.22, was identified as a potential therapeutic candidate. Normally OC90 is expressed in the cochlea with no known function in other normal tissues. In silico analysis of The Cancer Genome Atlas (TCGA) multi-tumor RNAseq cohorts revealed that OC90 is expressed in many tumor types at high prevalence and genomic amplification is associated with the elevated mRNA expression. In vitro assays in TNBC cell lines revealed OC90 expression with control over cell viability, apoptosis and invasion. RNA-seq analysis of OC90-siRNA knockdown and OC90-overexpression in BT20, BT549, HCC38 cell lines identified co-expressed transcripts, HMGA2, POLE2 and TRIB3. Altered expression of HMGA2, POLE2 and TRIB3 was predictive of survival among members of the Metabric breast cancer cohort. Thus, OC90 represents a potential therapeutic target whose knockdown could improve the treatment of TNBC.


Asunto(s)
Expresión Génica Ectópica , Glicoproteínas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Apoptosis , Western Blotting , Proteínas de Unión al Calcio , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias de la Mama Triple Negativas/metabolismo
15.
J Mol Diagn ; 21(1): 49-57, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30553750

RESUMEN

Prostate cancer is the most commonly diagnosed male cancer and the second leading cause of cancer deaths among men in the United States, with approximately 220,000 new diagnoses and approximately 27,000 deaths each year. Men with clinical low-risk disease can receive active surveillance to safely preserve quality of life, provided that the risk of an undetected aggressive cancer can be managed. Thus, prediction of a tumor's metastatic potential, ideally using only a biopsy sample, is critical to choosing appropriate treatment. We previously proposed and verified a metastasis potential score (MPS) based on regions prone to copy number alterations in metastatic prostate cancer; MPS is highly predictive of metastatic potential in primary tumors. We developed a novel, targeted postligation amplification sequencing approach, which we call the next-generation copy number alteration assay, to efficiently interrogate 902 genomic sites that belong to 194 genomic regions used in the MPS calculation. The assay is designed to work with the latest generation of sequencing platforms to produce estimates of copy number alteration events. The assay's technical reproducibility, robustness to low starting genomic material, and accuracy have been verified. The assay performed very well on cell lines, a cohort of prostate cancer surgical research samples, and matched punched biopsy samples, making it a significant step toward incorporating sequencing techniques for prostate cancer evaluation.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Neoplasias de la Próstata/genética , Variaciones en el Número de Copia de ADN , Pruebas Genéticas/economía , Pruebas Genéticas/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Masculino , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/etiología , Calidad de Vida , Factores de Riesgo , Factores de Tiempo
16.
Genes Cancer ; 9(1-2): 66-77, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29725504

RESUMEN

Copy number alterations(CNAs) are the most common genetic changes observed in many cancers, reflecting the innate chromosomal instability of this disorder. Yet, how these alterations affect gene function to promote metastases across different tumor types has not been established. In this study, we developed a pan-cancer metastasis potential score (panMPS) based on observed CNAs. panMPS predicts metastasis and metastasis-free survival in cohorts of patients with prostate cancer, triple negative breast cancer and lung adenocarcinoma, and overall survival in the Metabric breast cancer cohort and three cohorts from The Cancer Genome Atlas (TCGA), including prostate, breast and lung adenocarcinoma. These CNAs are present in cell lines of metastatic tumors from eight different origins, reflected by an elevated panMPS for all cell lines. Many copy number alterations involve large chromosomal segments that encompass multiple genes ("clumps"). We show that harnessing this structural information to select only one gene per clump captures the contributions of other genes within the clump, resulting in a robust predictor of metastasis outcome. These sets of selected genes are distinct from cancer drivers that undergo mutation, and in fact, metastasis-related functions have been published for over half of them.

17.
Genet Med ; 19(9): 1071-1077, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28301456

RESUMEN

PURPOSE: Identifying women at high risk for breast cancer can trigger a personal program of annual mammograms and magnetic resonance imaging scans for early detection, prophylactic surgery, or chemoprevention to reduce the risk of cancer. Yet, current strategies to identify high-risk mutations based on sequencing panels of genes have significant false-positive and false-negative results, suggesting the need for alternative approaches. METHODS: Flow-variant assays (FVAs) that assess the effects of mutations in the double-strand break (DSB) repair genetic pathway in lymphoblastoid cells in response to treatment with radiomimetic agents were assessed for sensitivity, specificity, and accuracy both alone and as part of a logistic regression classification score. In turn, these assays were validated in circulating B cells and applied to individuals with personal and/or family history of breast and/or ovarian cancer. RESULTS: A three-FVA classification score based on logistic regression had 95% accuracy. Individuals from a breast cancer-positive cohort with affected family members had high-risk FVA classification scores. CONCLUSION: Application of a classification score based on multiple FVAs could represent an alternative to panel sequencing for identifying women at high risk for cancer.Genet Med advance online publication 16 March 2017.


Asunto(s)
Linfocitos B/metabolismo , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/genética , Mutación , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Biomarcadores de Tumor , Neoplasias de la Mama/diagnóstico , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Análisis Mutacional de ADN , Detección Precoz del Cáncer/métodos , Detección Precoz del Cáncer/normas , Femenino , Genes p53 , Humanos , Fosforilación , Transporte de Proteínas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Análisis de Secuencia de ADN , Secuenciación del Exoma , Secuenciación Completa del Genoma
18.
DNA Repair (Amst) ; 38: 94-101, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26704428

RESUMEN

This review discusses the role of DNA mismatch repair (MMR) in the DNA damage response (DDR) that triggers cell cycle arrest and, in some cases, apoptosis. Although the focus is on findings from mammalian cells, much has been learned from studies in other organisms including bacteria and yeast [1,2]. MMR promotes a DDR mediated by a key signaling kinase, ATM and Rad3-related (ATR), in response to various types of DNA damage including some encountered in widely used chemotherapy regimes. An introduction to the DDR mediated by ATR reveals its immense complexity and highlights the many biological and mechanistic questions that remain. Recent findings and future directions are highlighted.


Asunto(s)
Daño del ADN , Reparación de la Incompatibilidad de ADN , Animales , Aductos de ADN , Metilación de ADN , Humanos , Transducción de Señal
20.
Hum Mol Genet ; 24(11): 3030-7, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25652403

RESUMEN

Heritable mutations in the BRCA1 and BRCA2 and other genes in the DNA double-strand break (DSB) repair pathway disrupt binding of the encoded proteins, transport into the nucleus and initiation of homologous recombination, thereby increasing cancer risk [Scully, R., Chen, J., Plug, A., Xiao, Y., Weaver, D., Feunteun, J., Ashley, T. and Livingston, D.M. (1997) Association of BRCA1 with Rad51 in mitotic and meiotic cells. Cell, 88, 265-275, Chen, J., Silver, D.P., Walpita, D., Cantor, S.B., Gazdar, A.F., Tomlinson, G., Couch, F.J., Weber, B.L., Ashley, T., Livingston, D.M. et al. (1998) Stable interaction between the products of the BRCA1 and BRCA2 tumor suppressor genes in mitotic and meiotic cells. Mol. Cell, 2, 317-328]. To meet the challenge of correct classification, flow cytometry-based functional variant analyses (FVAs) were developed to determine whether variants in DSB repair genes disrupted the binding of BRCA1 to BARD1, PALB2, BRCA2 and FANCD2, phosphorylation of p53 or BRCA1 nuclear localization in response to DNA damage caused by diepoxybutane, mitomycin C and bleomycin. Lymphoblastoid cells from individuals with BRCA1 pathogenic mutations, benign variants, and variants of uncertain significance or with known BRCA2, FANCC or NBN mutations were tested. Mutations in BRCA1 decreased nuclear localization of BRCA1 in response to individual or combination drug treatment. Mutations in BRCA1 reduced binding to co-factors, PALB2 and FANCD2 and decreased phosphorylation of p53. Mutations in BRCA2, FANCC and NBN decreased nuclear localization of BRCA1 in response to drug treatment, cofactors binding and p53 phosphorylation. Unsupervised cluster analysis of all and as few as two assays demonstrated two apparent clusters, high-risk BRCA1 mutations and phenocopies and low-risk, fully sequenced controls and variants of uncertain significance (VUS). Thus, two FVA assays distinguish BRCA1 mutations and phenocopies from benign variants and categorize most VUS as benign. Mutations in other DSB repair pathway genes produce molecular phenocopies. FVA assays may represent an adjunct to sequencing for categorizing VUS or may represent a stand-alone measure for assessing breast cancer risk.


Asunto(s)
Proteína BRCA1/genética , Roturas del ADN de Doble Cadena , Reparación del ADN por Recombinación , Proteína BRCA1/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Humanos , Anotación de Secuencia Molecular , Mutación , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA