Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Nat Rev Clin Oncol ; 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38992122

RESUMEN

Chromosomal instability (CIN) is a hallmark of cancer and a driver of metastatic dissemination, therapeutic resistance, and immune evasion. CIN is present in 60-80% of human cancers and poses a formidable therapeutic challenge as evidenced by the lack of clinically approved drugs that directly target CIN. This limitation in part reflects a lack of well-defined druggable targets as well as a dearth of tractable biomarkers enabling direct assessment and quantification of CIN in patients with cancer. Over the past decade, however, our understanding of the cellular mechanisms and consequences of CIN has greatly expanded, revealing novel therapeutic strategies for the treatment of chromosomally unstable tumours as well as new methods of assessing the dynamic nature of chromosome segregation errors that define CIN. In this Review, we describe advances that have shaped our understanding of CIN from a translational perspective, highlighting both challenges and opportunities in the development of therapeutic interventions for patients with chromosomally unstable cancers.

2.
Clin Cancer Res ; 30(12): 2672-2683, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38502113

RESUMEN

PURPOSE: Targeted therapies have improved outcomes for patients with metastatic colorectal cancer, but their impact is limited by rapid emergence of resistance. We hypothesized that an understanding of the underlying genetic mechanisms and intrinsic tumor features that mediate resistance to therapy will guide new therapeutic strategies and ultimately allow the prevention of resistance. EXPERIMENTAL DESIGN: We assembled a series of 52 patients with paired pretreatment and progression samples who received therapy targeting EGFR (n = 17), BRAF V600E (n = 17), KRAS G12C (n = 15), or amplified HER2 (n = 3) to identify molecular and clinical factors associated with time on treatment (TOT). RESULTS: All patients stopped treatment for progression and TOT did not vary by oncogenic driver (P = 0.5). Baseline disease burden (≥3 vs. <3 sites, P = 0.02), the presence of hepatic metastases (P = 0.02), and gene amplification on baseline tissue (P = 0.03) were each associated with shorter TOT. We found evidence of chromosomal instability (CIN) at progression in patients with baseline MAPK pathway amplifications and those with acquired gene amplifications. At resistance, copy-number changes (P = 0.008) and high number (≥5) of acquired alterations (P = 0.04) were associated with shorter TOT. Patients with hepatic metastases demonstrated both higher number of emergent alterations at resistance and enrichment of mutations involving receptor tyrosine kinases. CONCLUSIONS: Our genomic analysis suggests that high baseline CIN or effective induction of enhanced mutagenesis on targeted therapy underlies rapid progression. Longer response appears to result from a progressive acquisition of genomic or chromosomal instability in the underlying cancer or from the chance event of a new resistance alteration.


Asunto(s)
Neoplasias Colorrectales , Resistencia a Antineoplásicos , Terapia Molecular Dirigida , Proteínas Proto-Oncogénicas B-raf , Humanos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Femenino , Masculino , Proteínas Proto-Oncogénicas B-raf/genética , Persona de Mediana Edad , Anciano , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Mutación , Progresión de la Enfermedad , Receptores ErbB/genética , Receptores ErbB/antagonistas & inhibidores , Adulto , Inestabilidad Cromosómica , Anciano de 80 o más Años , Amplificación de Genes
3.
Cancer Immunol Res ; 12(6): 673-686, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38408184

RESUMEN

Chromosomal instability is a hallmark of human cancer that is associated with aggressive disease characteristics. Chromosome mis-segregations help fuel natural selection, but they risk provoking a cGAS-STING immune response through the accumulation of cytosolic DNA. The mechanisms of how tumors benefit from chromosomal instability while mitigating associated risks, such as enhanced immune surveillance, are poorly understood. Here, we identify cGAS-STING-dependent upregulation of the nuclease TREX1 as an adaptive, negative feedback mechanism that promotes immune evasion through digestion of cytosolic DNA. TREX1 loss diminishes tumor growth, prolongs survival of host animals, increases tumor immune infiltration, and potentiates response to immune checkpoint blockade selectively in tumors capable of mounting a type I IFN response downstream of STING. Together, these data demonstrate that TREX1 induction shields chromosomally unstable tumors from immune surveillance by dampening type I IFN production and suggest that TREX1 inhibitors might be used to selectively target tumors that have retained the inherent ability to mount an IFN response downstream of STING. See related article by Lim et al., p. 663.


Asunto(s)
Exodesoxirribonucleasas , Interferón Tipo I , Fosfoproteínas , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Interferón Tipo I/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Animales , Humanos , Ratones , Neoplasias/inmunología , Neoplasias/genética , Evasión Inmune , Línea Celular Tumoral , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Escape del Tumor
4.
Nat Cancer ; 5(3): 433-447, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38286827

RESUMEN

Liver metastasis (LM) confers poor survival and therapy resistance across cancer types, but the mechanisms of liver-metastatic organotropism remain unknown. Here, through in vivo CRISPR-Cas9 screens, we found that Pip4k2c loss conferred LM but had no impact on lung metastasis or primary tumor growth. Pip4k2c-deficient cells were hypersensitized to insulin-mediated PI3K/AKT signaling and exploited the insulin-rich liver milieu for organ-specific metastasis. We observed concordant changes in PIP4K2C expression and distinct metabolic changes in 3,511 patient melanomas, including primary tumors, LMs and lung metastases. We found that systemic PI3K inhibition exacerbated LM burden in mice injected with Pip4k2c-deficient cancer cells through host-mediated increase in hepatic insulin levels; however, this circuit could be broken by concurrent administration of an SGLT2 inhibitor or feeding of a ketogenic diet. Thus, this work demonstrates a rare example of metastatic organotropism through co-optation of physiological metabolic cues and proposes therapeutic avenues to counteract these mechanisms.


Asunto(s)
Neoplasias Hepáticas , Proteínas Proto-Oncogénicas c-akt , Humanos , Ratones , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas , Transducción de Señal , Insulina , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
5.
Cancer Discov ; 14(2): 214-226, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38197599

RESUMEN

Chromosome-containing micronuclei are a feature of human cancer. Micronuclei arise from chromosome mis-segregation and characterize tumors with elevated rates of chromosomal instability. Although their association with cancer has been long recognized, only recently have we broadened our understanding of the mechanisms that govern micronuclei formation and their role in tumor progression. In this review, we provide a brief historical account of micronuclei, depict the mechanisms underpinning their creation, and illuminate their capacity to propel tumor evolution through genetic, epigenetic, and transcriptional transformations. We also posit the prospect of leveraging micronuclei as biomarkers and therapeutic targets in chromosomally unstable cancers. SIGNIFICANCE: Micronuclei in chromosomally unstable cancer cells serve as pivotal catalysts for cancer progression, instigating transformative genomic, epigenetic, and transcriptional alterations. This comprehensive review not only synthesizes our present comprehension but also outlines a framework for translating this knowledge into pioneering biomarkers and therapeutics, thereby illuminating novel paths for personalized cancer management.


Asunto(s)
Neoplasias , Humanos , Neoplasias/genética , Inestabilidad Cromosómica , Segregación Cromosómica , Biomarcadores
6.
Mol Cell Oncol ; 10(1): 2014734, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38116246

RESUMEN

Our recent study revealed that APOBEC3B is upregulated during the preinvasive stages of non-small cell lung cancer and breast cancer. In addition to its role in mediating single nucleotide variants, we propose that APOBEC3 promotes copy number intratumor heterogeneity prior to invasion, providing a substrate for cancer evolution.

7.
Nature ; 620(7976): 1080-1088, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37612508

RESUMEN

Chromosomal instability (CIN) is a driver of cancer metastasis1-4, yet the extent to which this effect depends on the immune system remains unknown. Using ContactTracing-a newly developed, validated and benchmarked tool to infer the nature and conditional dependence of cell-cell interactions from single-cell transcriptomic data-we show that CIN-induced chronic activation of the cGAS-STING pathway promotes downstream signal re-wiring in cancer cells, leading to a pro-metastatic tumour microenvironment. This re-wiring is manifested by type I interferon tachyphylaxis selectively downstream of STING and a corresponding increase in cancer cell-derived endoplasmic reticulum (ER) stress response. Reversal of CIN, depletion of cancer cell STING or inhibition of ER stress response signalling abrogates CIN-dependent effects on the tumour microenvironment and suppresses metastasis in immune competent, but not severely immune compromised, settings. Treatment with STING inhibitors reduces CIN-driven metastasis in melanoma, breast and colorectal cancers in a manner dependent on tumour cell-intrinsic STING. Finally, we show that CIN and pervasive cGAS activation in micronuclei are associated with ER stress signalling, immune suppression and metastasis in human triple-negative breast cancer, highlighting a viable strategy to identify and therapeutically intervene in tumours spurred by CIN-induced inflammation.


Asunto(s)
Inestabilidad Cromosómica , Progresión de la Enfermedad , Neoplasias , Humanos , Benchmarking , Comunicación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Microambiente Tumoral , Interferón Tipo I/inmunología , Metástasis de la Neoplasia , Estrés del Retículo Endoplásmico , Transducción de Señal , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología
8.
Nature ; 619(7968): 176-183, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37286593

RESUMEN

Chromosomal instability (CIN) and epigenetic alterations are characteristics of advanced and metastatic cancers1-4, but whether they are mechanistically linked is unknown. Here we show that missegregation of mitotic chromosomes, their sequestration in micronuclei5,6 and subsequent rupture of the micronuclear envelope7 profoundly disrupt normal histone post-translational modifications (PTMs), a phenomenon conserved across humans and mice, as well as in cancer and non-transformed cells. Some of the changes in histone PTMs occur because of the rupture of the micronuclear envelope, whereas others are inherited from mitotic abnormalities before the micronucleus is formed. Using orthogonal approaches, we demonstrate that micronuclei exhibit extensive differences in chromatin accessibility, with a strong positional bias between promoters and distal or intergenic regions, in line with observed redistributions of histone PTMs. Inducing CIN causes widespread epigenetic dysregulation, and chromosomes that transit in micronuclei experience heritable abnormalities in their accessibility long after they have been reincorporated into the primary nucleus. Thus, as well as altering genomic copy number, CIN promotes epigenetic reprogramming and heterogeneity in cancer.


Asunto(s)
Inestabilidad Cromosómica , Segregación Cromosómica , Cromosomas , Epigénesis Genética , Micronúcleos con Defecto Cromosómico , Neoplasias , Animales , Humanos , Ratones , Cromatina/genética , Inestabilidad Cromosómica/genética , Cromosomas/genética , Cromosomas/metabolismo , Histonas/química , Histonas/metabolismo , Neoplasias/genética , Neoplasias/patología , Mitosis , Variaciones en el Número de Copia de ADN , Procesamiento Proteico-Postraduccional
9.
Cancer Cell ; 41(7): 1207-1221.e12, 2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37327789

RESUMEN

The cell-autonomous balance of immune-inhibitory and -stimulatory signals is a critical process in cancer immune evasion. Using patient-derived co-cultures, humanized mouse models, and single-cell RNA-sequencing of patient melanomas biopsied before and on immune checkpoint blockade, we find that intact cancer cell-intrinsic expression of CD58 and ligation to CD2 is required for anti-tumor immunity and is predictive of treatment response. Defects in this axis promote immune evasion through diminished T cell activation, impaired intratumoral T cell infiltration and proliferation, and concurrently increased PD-L1 protein stabilization. Through CRISPR-Cas9 and proteomics screens, we identify and validate CMTM6 as critical for CD58 stability and upregulation of PD-L1 upon CD58 loss. Competition between CD58 and PD-L1 for CMTM6 binding determines their rate of endosomal recycling over lysosomal degradation. Overall, we describe an underappreciated yet critical axis of cancer immunity and provide a molecular basis for how cancer cells balance immune inhibitory and stimulatory cues.


Asunto(s)
Antígeno B7-H1 , Melanoma , Ratones , Animales , Antígeno B7-H1/genética , Linfocitos T , Antígenos CD58/química , Antígenos CD58/metabolismo , Melanoma/genética , Melanoma/metabolismo , Activación de Linfocitos
10.
Nature ; 618(7967): 1041-1048, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37165191

RESUMEN

Complex genome rearrangements can be generated by the catastrophic pulverization of missegregated chromosomes trapped within micronuclei through a process known as chromothripsis1-5. As each chromosome contains a single centromere, it remains unclear how acentric fragments derived from shattered chromosomes are inherited between daughter cells during mitosis6. Here we tracked micronucleated chromosomes with live-cell imaging and show that acentric fragments cluster in close spatial proximity throughout mitosis for asymmetric inheritance by a single daughter cell. Mechanistically, the CIP2A-TOPBP1 complex prematurely associates with DNA lesions within ruptured micronuclei during interphase, which poises pulverized chromosomes for clustering upon mitotic entry. Inactivation of CIP2A-TOPBP1 caused acentric fragments to disperse throughout the mitotic cytoplasm, stochastically partition into the nucleus of both daughter cells and aberrantly misaccumulate as cytoplasmic DNA. Mitotic clustering facilitates the reassembly of acentric fragments into rearranged chromosomes lacking the extensive DNA copy-number losses that are characteristic of canonical chromothripsis. Comprehensive analysis of pan-cancer genomes revealed clusters of DNA copy-number-neutral rearrangements-termed balanced chromothripsis-across diverse tumour types resulting in the acquisition of known cancer driver events. Thus, distinct patterns of chromothripsis can be explained by the spatial clustering of pulverized chromosomes from micronuclei.


Asunto(s)
Cromosomas Humanos , Cromotripsis , Micronúcleos con Defecto Cromosómico , Mitosis , Humanos , Centrómero , Cromosomas Humanos/genética , ADN/genética , ADN/metabolismo , Variaciones en el Número de Copia de ADN , Interfase , Mitosis/genética , Neoplasias/genética
11.
Nat Mater ; 22(5): 532-533, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37138010
12.
Science ; 380(6640): 47, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37023189

RESUMEN

Journey through basic biology reveals a way to treat chromosomally unstable cancers.


Asunto(s)
Inestabilidad Cromosómica , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética
13.
J Exp Med ; 220(3)2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36534085

RESUMEN

Late cardiac toxicity is a potentially lethal complication of cancer therapy, yet the pathogenic mechanism remains largely unknown, and few treatment options exist. Here we report DNA-damaging agents such as radiation and anthracycline chemotherapies inducing delayed cardiac inflammation following therapy due to activation of cGAS- and STING-dependent type I interferon signaling. Genetic ablation of cGAS-STING signaling in mice inhibits DNA damage-induced cardiac inflammation, rescues late cardiac functional decline, and prevents death from cardiac events. Treatment with a STING antagonist suppresses cardiac interferon signaling following DNA-damaging therapies and effectively mitigates cardiac toxicity. These results identify a therapeutically targetable, pathogenic mechanism for one of the most vexing treatment-related toxicities in cancer survivors.


Asunto(s)
Antineoplásicos , Cardiotoxicidad , Daño del ADN , Neoplasias , Animales , Ratones , Inmunidad Innata , Inflamación , Neoplasias/tratamiento farmacológico , Nucleotidiltransferasas/genética , Antineoplásicos/efectos adversos
14.
Trends Cancer ; 8(10): 788-789, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35915014

RESUMEN

Chromosomal instability (CIN) is a key genomic driver of human cancer. CIN generates genomic copy-number heterogeneity and tumor-derived inflammation. In a recent paper, Hong et al. identify the cGAS-STING innate immune pathway as a crucial dependency in cancer cells with CIN and pinpoint the IL6/STAT3 axis as a therapeutic vulnerability in these difficult-to-treat tumors.


Asunto(s)
Interleucina-6 , Neoplasias , Inestabilidad Cromosómica/genética , Humanos , Inflamación/genética , Interleucina-6/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo
15.
Cell ; 185(14): 2591-2608.e30, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35803246

RESUMEN

Melanoma brain metastasis (MBM) frequently occurs in patients with advanced melanoma; yet, our understanding of the underlying salient biology is rudimentary. Here, we performed single-cell/nucleus RNA-seq in 22 treatment-naive MBMs and 10 extracranial melanoma metastases (ECMs) and matched spatial single-cell transcriptomics and T cell receptor (TCR)-seq. Cancer cells from MBM were more chromosomally unstable, adopted a neuronal-like cell state, and enriched for spatially variably expressed metabolic pathways. Key observations were validated in independent patient cohorts, patient-derived MBM/ECM xenograft models, RNA/ATAC-seq, proteomics, and multiplexed imaging. Integrated spatial analyses revealed distinct geography of putative cancer immune evasion and evidence for more abundant intra-tumoral B to plasma cell differentiation in lymphoid aggregates in MBM. MBM harbored larger fractions of monocyte-derived macrophages and dysfunctional TOX+CD8+ T cells with distinct expression of immune checkpoints. This work provides comprehensive insights into MBM biology and serves as a foundational resource for further discovery and therapeutic exploration.


Asunto(s)
Neoplasias Encefálicas , Melanoma , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/secundario , Linfocitos T CD8-positivos/patología , Ecosistema , Humanos , RNA-Seq
16.
Curr Opin Genet Dev ; 74: 101913, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35526333

RESUMEN

Chromosomal instability (CIN) is a hallmark of the most aggressive malignancies. Features of these tumors include complex genomic rearrangements, the presence of mis-segregated chromosomes in micronuclei, and extrachromosomal DNA (ecDNA) formation. Here, we review the development of CIN, and examine CIN in the context of cancer evolution, tumor genomic evolution, and therapeutic resistance. We also discuss the role of whole-genome duplications, breakage-fusion-bridge cycles, ecDNA or double minutes in gene amplification promoting tumor evolution.


Asunto(s)
Inestabilidad Cromosómica , Neoplasias , Inestabilidad Cromosómica/genética , Amplificación de Genes , Inestabilidad Genómica , Genómica , Humanos , Neoplasias/patología , Oncogenes
17.
J Mol Cell Biol ; 14(4)2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35325182

RESUMEN

Cytosolic DNA is prevalent in cells constituting the tumor microenvironment (TME) and can activate the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) innate immune pathway. The initiation, transmission, and execution of the cGAS-STING pathway can take place among different cell types within the TME and thus cGAS-STING may play opposing roles in driving tumor progression in addition to its tumor cell-intrinsic role. Herein, we review recent advances in the cGAS-STING field with a focus on its crosstalk with other signaling pathways in the TME. Future efforts to depict a more detailed picture of the roles of cGAS-STING signaling in the TME will help design a better cancer treatment regime by targeting the cGAS-STING pathway more precisely.


Asunto(s)
Proteínas de la Membrana , Microambiente Tumoral , Citosol/metabolismo , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal
19.
Cell Rep Med ; 2(9): 100406, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34622240

RESUMEN

In the current issue of Cancer Cell, Lan et al.1 demonstrate that a bifunctional fusion protein targeting TGF-ß and PD-L1 can synergize with radiation therapy to simultaneously augment tumor control and reduce normal tissue toxicity.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Humanos , Neoplasias/radioterapia , Factor de Crecimiento Transformador beta
20.
Nat Commun ; 12(1): 5402, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34518527

RESUMEN

Chromosomal instability (CIN) and epigenetic alterations have been implicated in tumor progression and metastasis; yet how these two hallmarks of cancer are related remains poorly understood. By integrating genetic, epigenetic, and functional analyses at the single cell level, we show that progression of uveal melanoma (UM), the most common intraocular primary cancer in adults, is driven by loss of Polycomb Repressive Complex 1 (PRC1) in a subpopulation of tumor cells. This leads to transcriptional de-repression of PRC1-target genes and mitotic chromosome segregation errors. Ensuing CIN leads to the formation of rupture-prone micronuclei, exposing genomic double-stranded DNA (dsDNA) to the cytosol. This provokes tumor cell-intrinsic inflammatory signaling, mediated by aberrant activation of the cGAS-STING pathway. PRC1 inhibition promotes nuclear enlargement, induces a transcriptional response that is associated with significantly worse patient survival and clinical outcomes, and enhances migration that is rescued upon pharmacologic inhibition of CIN or STING. Thus, deregulation of PRC1 can promote tumor progression by inducing CIN and represents an opportunity for early therapeutic intervention.


Asunto(s)
Inestabilidad Cromosómica , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Complejo Represivo Polycomb 1/genética , Neoplasias de la Úvea/genética , Línea Celular Tumoral , Segregación Cromosómica/genética , Progresión de la Enfermedad , Células HEK293 , Humanos , Melanoma/metabolismo , Melanoma/patología , Complejo Represivo Polycomb 1/metabolismo , Pronóstico , RNA-Seq/métodos , Transducción de Señal/genética , Análisis de Supervivencia , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...