Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Autoimmun ; 99: 24-32, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30679006

RESUMEN

Systematic Lupus Erythematosus (SLE) is an autoimmune syndrome of unclear etiology. While T and B cell abnormalities contribute to disease pathogenesis, recent work suggests that inflammatory M1-like macrophages also play a role. Previous work showed that the TLR2/1 agonist PAM3CSK4 (PAM3) could stimulate normal human monocytes to preferentially differentiate into immunosuppressive M2-like rather than inflammatory M1-like macrophages. This raised the possibility of PAM3 being used to normalize the M1:M2 ratio in SLE. Consistent with that possibility, monocytes from lupus patients differentiated into M2-like macrophages when treated with PAM3 in vitro. Furthermore, lupus-prone NZB x NZW F1 mice responded similarly to weekly PAM3 treatment. Normalization of the M2 macrophage frequency was associated with delayed disease progression, decreased autoantibody and inflammatory cytokine synthesis, reduced proteinuria and prolonged survival in NZB x NZW F1 mice. The ability of PAM3 to bias monocyte differentiation in favor of immunosuppressive macrophages may represent a novel approach to the therapy of SLE.


Asunto(s)
Lipopéptidos/farmacología , Lupus Eritematoso Sistémico/etiología , Lupus Eritematoso Sistémico/metabolismo , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Animales , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/inmunología , Citocinas/metabolismo , Endocitosis/inmunología , Femenino , Inmunofenotipificación , Lupus Eritematoso Sistémico/complicaciones , Nefritis Lúpica/etiología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Macrófagos/metabolismo , Masculino , Ratones , Monocitos/metabolismo , Receptor Toll-Like 1/metabolismo , Receptor Toll-Like 2/metabolismo
2.
J Immunol ; 194(9): 4215-21, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25825448

RESUMEN

Tumors persist by occupying immunosuppressive microenvironments that inhibit the activity of tumoricidal T and NK cells. Monocytic myeloid-derived suppressor cells (mMDSC) are an important component of this immunosuppressive milieu. We find that the suppressive activity of mMDSC isolated from cancer patients can be reversed by treatment with TLR7/8 agonists, which induce human mMDSC to differentiate into tumoricidal M1-like macrophages. In contrast, agonists targeting TLR1/2 cause mMDSC to mature into immunosuppressive M2-like macrophages. These two populations of macrophage are phenotypically and functionally discrete and differ in gene expression profile. The ability of TLR7/8 agonists to reverse mMDSC-mediated immune suppression suggests that they might be useful adjuncts for tumor immunotherapy.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Imidazoles/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , Células Mieloides/citología , Células Mieloides/efectos de los fármacos , Receptores Toll-Like/agonistas , Humanos , Tolerancia Inmunológica/inmunología , Monocitos/inmunología , Células Mieloides/inmunología , Relación Estructura-Actividad , Receptores Toll-Like/inmunología
3.
Pharmacol Res ; 105: 216-25, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26779666

RESUMEN

Synthetic oligodeoxynucleotides that can down-regulate cellular elements of the immune system have been developed and are being widely studied in preclinical models. These agents vary in sequence, mechanism of action, and cellular target(s) but share the ability to suppress a plethora of inflammatory responses. This work reviews the types of immunosuppressive oligodeoxynucleotide (Sup ODN) and compares their therapeutic activity against diseases characterized by pathologic levels of immune stimulation ranging from autoimmunity to septic shock to cancer (see graphical abstract). The mechanism(s) underlying the efficacy of Sup ODN and the influence size, sequence and nucleotide backbone on function are considered.


Asunto(s)
Inmunosupresores/química , Inmunosupresores/uso terapéutico , Oligodesoxirribonucleótidos/química , Oligodesoxirribonucleótidos/uso terapéutico , Animales , Secuencia de Bases , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Inmunosupresores/farmacología , Factores Reguladores del Interferón/inmunología , Oligodesoxirribonucleótidos/farmacología , Receptor Toll-Like 9/inmunología
4.
Pathog Immun ; 9(1): 56-90, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690563

RESUMEN

There are a growing number of studies linking the composition of the human microbiome to disease states and treatment responses, especially in the context of cancer. This has raised significant interest in developing microbes and microbial products as cancer immunotherapeutics that mimic or recapitulate the beneficial effects of host-microbe interactions. Bacterial extracellular vesicles (bEVs) are nano-sized, membrane-bound particles secreted by essentially all bacteria species and contain a diverse bioactive cargo of the producing cell. They have a fundamental role in facilitating interactions among cells of the same species, different microbial species, and even with multicellular host organisms in the context of colonization (microbiome) and infection. The interaction of bEVs with the immune system has been studied extensively in the context of infection and suggests that bEV effects depend largely on the producing species. They thus provide functional diversity, while also being nonreplicative, having inherent cell-targeting qualities, and potentially overcoming natural barriers. These characteristics make them highly appealing for development as cancer immunotherapeutics. Both natively secreted and engineered bEVs are now being investigated for their application as immunotherapeutics, vaccines, drug delivery vehicles, and combinations of the above, with promising early results. This suggests that both the intrinsic immunomodulatory properties of bEVs and their ability to be modified could be harnessed for the development of next-generation microbe-inspired therapies. Nonetheless, there remain major outstanding questions regarding how the observed preclinical effectiveness will translate from murine models to primates, and humans in particular. Moreover, research into the pharmacology, toxicology, and mass manufacturing of this potential novel therapeutic platform is still at early stages. In this review, we highlight the breadth of bEV interactions with host cells, focusing on immunologic effects as the main mechanism of action of bEVs currently in preclinical development. We review the literature on ongoing efforts to develop natively secreted and engineered bEVs from a variety of bacterial species for cancer therapy and finally discuss efforts to overcome outstanding challenges that remain for clinical translation.

5.
Trends Cancer ; 10(1): 15-27, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37625928

RESUMEN

The Swiss cheese model is used to assess risks and explain accidents in a variety of industries. This model can be applied to dissect the homeostatic mechanisms whose cumulative dysregulation contributes to disease states, including cancer. Using glioblastoma (GBM) as an exemplar, we discuss how specific protumorigenic mechanisms collectively drive disease by affecting genomic integrity, epigenetic regulation, metabolic homeostasis, and antitumor immunity. We further highlight how host factors, such as hormonal differences and aging, impact this process, and the interplay between these 'system failures' that enable tumor progression and foster therapeutic resistance. Finally, we examine therapies that consider the interactions between these elements, which may comprise more effective approaches given the multifaceted protumorigenic mechanisms that drive GBM.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/patología , Epigénesis Genética
6.
Cell Rep Med ; 4(11): 101293, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37992680

RESUMEN

In this issue, Pang and colleagues1 identify the protease legumain as a potential immunotherapy target in glioblastoma that drives tumor-associated macrophages in response to hypoxia.


Asunto(s)
Glioblastoma , Humanos , Cisteína Endopeptidasas/fisiología , Terapia de Inmunosupresión/efectos adversos , Hipoxia
7.
medRxiv ; 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36945620

RESUMEN

Glioblastoma (GBM) treatment includes maximal safe resection of the core and MRI contrast-enhancing (CE) tumor. Complete resection of the infiltrative non-contrast-enhancing (NCE) tumor rim is rarely achieved. We established a safe, semi-automated workflow for spatially-registered sampling of MRI-defined GBM regions in 19 patients with downstream analysis and biobanking, enabling studies of NCE, wherefrom recurrence/progression typically occurs. Immunophenotyping revealed underrepresentation of myeloid cell subsets and CD8+ T cells in the NCE. While NCE T cells phenotypically and functionally resembled those in matching CE tumor, subsets of activated (CD69hi) effector memory CD8+ T cells were overrepresented. Contrarily, CD25hi Tregs and other subsets were underrepresented. Overall, our study demonstrated that MRI-guided, spatially-registered, intraoperative immunosampling is feasible as part of routine GBM surgery. Further elucidation of the shared and spatially distinct microenvironmental biology of GBM will enable development of therapeutic approaches targeting the NCE infiltrative tumor to decrease GBM recurrence.

8.
Elife ; 122023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36876914

RESUMEN

Recent studies reveal that lateral mitochondrial transfer, the movement of mitochondria from one cell to another, can affect cellular and tissue homeostasis. Most of what we know about mitochondrial transfer stems from bulk cell studies and have led to the paradigm that functional transferred mitochondria restore bioenergetics and revitalize cellular functions to recipient cells with damaged or non-functional mitochondrial networks. However, we show that mitochondrial transfer also occurs between cells with functioning endogenous mitochondrial networks, but the mechanisms underlying how transferred mitochondria can promote such sustained behavioral reprogramming remain unclear. We report that unexpectedly, transferred macrophage mitochondria are dysfunctional and accumulate reactive oxygen species in recipient cancer cells. We further discovered that reactive oxygen species accumulation activates ERK signaling, promoting cancer cell proliferation. Pro-tumorigenic macrophages exhibit fragmented mitochondrial networks, leading to higher rates of mitochondrial transfer to cancer cells. Finally, we observe that macrophage mitochondrial transfer promotes tumor cell proliferation in vivo. Collectively these results indicate that transferred macrophage mitochondria activate downstream signaling pathways in a ROS-dependent manner in cancer cells, and provide a model of how sustained behavioral reprogramming can be mediated by a relatively small amount of transferred mitochondria in vitro and in vivo.


Asunto(s)
Mitocondrias , Neoplasias , Humanos , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/metabolismo , Neoplasias/patología , Transducción de Señal , Proliferación Celular
9.
Cancer Discov ; 13(9): 2090-2105, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37378557

RESUMEN

Sex differences in glioblastoma (GBM) incidence and outcome are well recognized, and emerging evidence suggests that these extend to genetic/epigenetic and cellular differences, including immune responses. However, the mechanisms driving immunologic sex differences are not fully understood. Here, we demonstrate that T cells play a critical role in driving GBM sex differences. Male mice exhibited accelerated tumor growth, with decreased frequency and increased exhaustion of CD8+ T cells in the tumor. Furthermore, a higher frequency of progenitor exhausted T cells was found in males, with improved responsiveness to anti-PD-1 treatment. Moreover, increased T-cell exhaustion was observed in male GBM patients. Bone marrow chimera and adoptive transfer models indicated that T cell-mediated tumor control was predominantly regulated in a cell-intrinsic manner, partially mediated by the X chromosome inactivation escape gene Kdm6a. These findings demonstrate that sex-biased predetermined behavior of T cells is critical for inducing sex differences in GBM progression and immunotherapy response. SIGNIFICANCE: Immunotherapies in patients with GBM have been unsuccessful due to a variety of factors, including the highly immunosuppressive tumor microenvironment in GBM. This study demonstrates that sex-biased T-cell behaviors are predominantly intrinsically regulated, further suggesting sex-specific approaches can be leveraged to potentially improve the therapeutic efficacy of immunotherapy in GBM. See related commentary by Alspach, p. 1966. This article is featured in Selected Articles from This Issue, p. 1949.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Masculino , Femenino , Ratones , Animales , Glioblastoma/genética , Agotamiento de Células T , Linfocitos T CD8-positivos , Inmunoterapia , Inmunidad , Neoplasias Encefálicas/patología , Microambiente Tumoral
10.
bioRxiv ; 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-38014234

RESUMEN

The glioblastoma microenvironment is enriched in immunosuppressive factors that potently interfere with the function of cytotoxic T lymphocytes. Cancer cells can directly impact the immune system, but the mechanisms driving these interactions are not completely clear. Here we demonstrate that the polyamine metabolite spermidine is elevated in the glioblastoma tumor microenvironment. Exogenous administration of spermidine drives tumor aggressiveness in an immune-dependent manner in pre-clinical mouse models via reduction of CD8+ T cell frequency and phenotype. Knockdown of ornithine decarboxylase, the rate-limiting enzyme in spermidine synthesis, did not impact cancer cell growth in vitro but did result in extended survival. Furthermore, glioblastoma patients with a more favorable outcome had a significant reduction in spermidine compared to patients with a poor prognosis. Our results demonstrate that spermidine functions as a cancer cell-derived metabolite that drives tumor progression by reducing CD8+T cell number and function.

11.
JCI Insight ; 8(13)2023 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-37252795

RESUMEN

Intratumoral heterogeneity is a defining hallmark of glioblastoma, driving drug resistance and ultimately recurrence. Many somatic drivers of microenvironmental change have been shown to affect this heterogeneity and, ultimately, the treatment response. However, little is known about how germline mutations affect the tumoral microenvironment. Here, we find that the single-nucleotide polymorphism (SNP) rs755622 in the promoter of the cytokine macrophage migration inhibitory factor (MIF) is associated with increased leukocyte infiltration in glioblastoma. Furthermore, we identified an association between rs755622 and lactotransferrin expression, which could also be used as a biomarker for immune-infiltrated tumors. These findings demonstrate that a germline SNP in the promoter region of MIF may affect the immune microenvironment and further reveal a link between lactotransferrin and immune activation.


Asunto(s)
Glioblastoma , Factores Inhibidores de la Migración de Macrófagos , Humanos , Lactoferrina/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Polimorfismo de Nucleótido Simple , Glioblastoma/genética , Regiones Promotoras Genéticas , Microambiente Tumoral/genética , Oxidorreductasas Intramoleculares/genética
12.
Nat Cancer ; 4(5): 648-664, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37169842

RESUMEN

The transfer of intact mitochondria between heterogeneous cell types has been confirmed in various settings, including cancer. However, the functional implications of mitochondria transfer on tumor biology are poorly understood. Here we show that mitochondria transfer is a prevalent phenomenon in glioblastoma (GBM), the most frequent and malignant primary brain tumor. We identified horizontal mitochondria transfer from astrocytes as a mechanism that enhances tumorigenesis in GBM. This transfer is dependent on network-forming intercellular connections between GBM cells and astrocytes, which are facilitated by growth-associated protein 43 (GAP43), a protein involved in neuron axon regeneration and astrocyte reactivity. The acquisition of astrocyte mitochondria drives an increase in mitochondrial respiration and upregulation of metabolic pathways linked to proliferation and tumorigenicity. Functionally, uptake of astrocyte mitochondria promotes cell cycle progression to proliferative G2/M phases and enhances self-renewal and tumorigenicity of GBM. Collectively, our findings reveal a host-tumor interaction that drives proliferation and self-renewal of cancer cells, providing opportunities for therapeutic development.


Asunto(s)
Glioblastoma , Humanos , Astrocitos/metabolismo , Astrocitos/patología , Proteína GAP-43/metabolismo , Proteína GAP-43/uso terapéutico , Axones/metabolismo , Axones/patología , Línea Celular Tumoral , Regeneración Nerviosa , Mitocondrias/metabolismo , Mitocondrias/patología
13.
Cancer Immunol Res ; 10(7): 787, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35670711

RESUMEN

Lack of insight into the immunosuppressive microenvironment of glioblastoma (GBM) hinders successful application of immunotherapy. In this issue, Alanio and colleagues identify a distinct T-cell localization profile between recurrent and de novo GBM highlighting the importance of spatial heterogeneity and providing new avenues to explore to improve GBM outcome. See related article by Alanio et al., p. 800 (3).


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Humanos , Inmunoterapia , Linfocitos T/patología , Microambiente Tumoral/inmunología
14.
Exp Suppl ; 113: 189-217, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35165865

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are immature bone marrow-derived suppressive cells that are an important component of the pathological immune response associated with cancer. Expansion of MDSCs has been linked to poor disease outcome and therapeutic resistance in patients with various malignancies, making these cells potential targets for next-generation treatment strategies. MDSCs are classified into monocytic (M-MDSC) and polymorphonuclear/granulocytic (PMN-MDSC) subtypes that undertake distinct and numerous roles in the tumor microenvironment or systemically to drive disease progression. In this chapter, we will discuss how MDSC subsets contribute to the growth of primary tumors and induce metastatic spread by suppressing the antitumor immune response, supporting cancer stem cell (CSC)/epithelial-to-mesenchymal transition (EMT) phenotypes and promoting angiogenesis. We will also summarize the signaling networks involved in the crosstalk between cancer cells and MDSCs that could represent putative immunotherapy targets.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias , Humanos , Monocitos , Neoplasias/genética , Neovascularización Patológica , Microambiente Tumoral/genética
15.
Cancer Res ; 82(14): 2515-2516, 2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35844171

RESUMEN

Lung adenocarcinoma is the most common subtype of lung cancer, which has the second highest incidence among cancers. Immunotherapy has revolutionized lung cancer treatment, yet the checkpoint blockade response rate is less than 20% in patients with lung adenocarcinoma. As lung adenocarcinoma consists of heterogeneous histologic subsets with diverse tumor invasion phenotypes and clinical outcomes, understanding the mechanisms of resistance based on the histology subset is essential. In the current issue, Jang and colleagues demonstrated that PD-1-expressing macrophages are the dominant immune cell population in the tumor-immune microenvironment (TiME) of invasive lung adenocarcinoma and are responsible for driving tumor progression from preinvasive to invasive subtypes. PD-1-expressing macrophages are protumorigenic and highly plastic, potentially promoting invasive solid tumor development. Ablation of macrophages remodels the TiME and leads to a favorable anti-PD-1 blockade response, suggesting a potential combination therapy in patients with lung adenocarcinoma resistant to monotherapy. This current work highlights the spatiotemporal dynamics of the TiME during lung adenocarcinoma progression and the critical role of PD-1-expressing macrophages in driving tumorigenesis as well as resistance to immunotherapy. See related article by Jang et al., p. 2593.

16.
J Invest Dermatol ; 142(8): 2159-2172.e9, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35090950

RESUMEN

TNF-α, a proinflammatory cytokine, is a crucial mediator of psoriasis pathogenesis. TNF-α functions by activating TNFR1 and TNFR2. Anti-TNF drugs that neutralize TNF-α, thus blocking the activation of TNFR1 and TNFR2, have been proven highly therapeutic in psoriatic diseases. TNF-α also plays an important role in host defense; thus, anti-TNF therapy can cause potentially serious adverse effects, including opportunistic infections and latent tuberculosis reactivation. These adverse effects are attributed to TNFR1 inactivation. Therefore, understanding the relative contributions of TNFR1 and TNFR2 has clinical implications in mitigating psoriasis versus global TNF-α blockade. We found a significant reduction in psoriasis lesions as measured by epidermal hyperplasia, characteristic gross skin lesion, and IL-23 or IL-17A levels in Tnfr2-knockout but not in Tnfr1-knockout mice in the imiquimod psoriasis model. Furthermore, imiquimod-mediated increase in the myeloid dendritic cells, TNF/inducible nitric oxide synthase‒producing dendritic cells, and IL-23 expression in the draining lymph nodes were dependent on TNFR2 but not on TNFR1. Together, our results support that psoriatic inflammation is not dependent on TNFR1 activity but is driven by a TNFR2-dependent IL-23/IL-17 pathway activation. Thus, targeting the TNFR2 pathway may emerge as a potential next-generation therapeutic approach for psoriatic diseases.


Asunto(s)
Psoriasis , Receptores Tipo II del Factor de Necrosis Tumoral , Animales , Células Dendríticas/metabolismo , Imiquimod , Inflamación/patología , Interleucina-17 , Interleucina-23 , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Psoriasis/tratamiento farmacológico , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Inhibidores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/metabolismo
17.
Cell Rep ; 40(11): 111348, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36103817

RESUMEN

Despite therapeutic interventions for glioblastoma (GBM), cancer stem cells (CSCs) drive recurrence. The precise mechanisms underlying CSC resistance, namely inhibition of cell death, are unclear. We built on previous observations that the high cell surface expression of junctional adhesion molecule-A drives CSC maintenance and identified downstream signaling networks, including the cysteine protease inhibitor SerpinB3. Using genetic depletion approaches, we found that SerpinB3 is necessary for CSC maintenance, survival, and tumor growth, as well as CSC pathway activation. Knockdown of SerpinB3 also increased apoptosis and susceptibility to radiation therapy. SerpinB3 was essential to buffer cathepsin L-mediated cell death, which was enhanced with radiation. Finally, we found that SerpinB3 knockdown increased the efficacy of radiation in pre-clinical models. Taken together, our findings identify a GBM CSC-specific survival mechanism involving a cysteine protease inhibitor, SerpinB3, and provide a potential target to improve the efficacy of GBM therapies against therapeutically resistant CSCs.


Asunto(s)
Glioblastoma , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Glioblastoma/patología , Humanos , Células Madre Neoplásicas/metabolismo , Transducción de Señal
18.
Cancer Res ; 82(22): 4274-4287, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36126163

RESUMEN

In multiple types of cancer, an increased frequency in myeloid-derived suppressor cells (MDSC) is associated with worse outcomes and poor therapeutic response. In the glioblastoma (GBM) microenvironment, monocytic (m) MDSCs represent the predominant subset. However, the molecular basis of mMDSC enrichment in the tumor microenvironment compared with granulocytic (g) MDSCs has yet to be determined. Here we performed the first broad epigenetic profiling of MDSC subsets to define underlying cell-intrinsic differences in behavior and found that enhanced gene accessibility of cell adhesion programs in mMDSCs is linked to their tumor-accelerating ability in GBM models upon adoptive transfer. Mouse and human mMDSCs expressed higher levels of integrin ß1 and dipeptidyl peptidase-4 (DPP-4) compared with gMDSCs as part of an enhanced cell adhesion signature. Integrin ß1 blockade abrogated the tumor-promoting phenotype of mMDSCs and altered the immune profile in the tumor microenvironment, whereas treatment with a DPP-4 inhibitor extended survival in preclinical GBM models. Targeting DPP-4 in mMDSCs reduced pERK signaling and their migration towards tumor cells. These findings uncover a fundamental difference in the molecular basis of MDSC subsets and suggest that integrin ß1 and DPP-4 represent putative immunotherapy targets to attenuate myeloid cell-driven immune suppression in GBM. SIGNIFICANCE: Epigenetic profiling uncovers cell adhesion programming as a regulator of the tumor-promoting functions of monocytic myeloid-derived suppressor cells in glioblastoma, identifying therapeutic targets that modulate the immune response and suppress tumor growth.


Asunto(s)
Adhesión Celular , Glioblastoma , Células Supresoras de Origen Mieloide , Animales , Humanos , Ratones , Glioblastoma/metabolismo , Glioblastoma/patología , Integrina beta1/metabolismo , Células Supresoras de Origen Mieloide/patología , Microambiente Tumoral
19.
Cancer Res ; 82(24): 4654-4669, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36206317

RESUMEN

Epithelial ovarian cancer (EOC) is the leading cause of gynecologic cancer death. Despite initial responses to intervention, up to 80% of patient tumors recur and require additional treatment. Retrospective clinical analysis of patients with ovarian cancer indicates antibiotic use during chemotherapy treatment is associated with poor overall survival. Here, we assessed whether antibiotic (ABX) treatment would impact growth of EOC and sensitivity to cisplatin. Immunocompetent or immunocompromised mice were given untreated control or ABX-containing (metronidazole, ampicillin, vancomycin, and neomycin) water prior to intraperitoneal injection with EOC cells, and cisplatin therapy was administered biweekly until endpoint. Tumor-bearing ABX-treated mice exhibited accelerated tumor growth and resistance to cisplatin therapy compared with control treatment. ABX treatment led to reduced apoptosis, increased DNA damage repair, and enhanced angiogenesis in cisplatin-treated tumors, and tumors from ABX-treated mice contained a higher frequency of cisplatin-augmented cancer stem cells than control mice. Stool analysis indicated nonresistant gut microbial species were disrupted by ABX treatment. Cecal transplants of microbiota derived from control-treated mice was sufficient to ameliorate chemoresistance and prolong survival of ABX-treated mice, indicative of a gut-derived tumor suppressor. Metabolomics analyses identified circulating gut-derived metabolites that were altered by ABX treatment and restored by recolonization, providing candidate metabolites that mediate the cross-talk between the gut microbiome and ovarian cancer. Collectively, these findings indicate that an intact microbiome functions as a tumor suppressor in EOC, and perturbation of the gut microbiota with ABX treatment promotes tumor growth and suppresses cisplatin sensitivity. SIGNIFICANCE: Restoration of the gut microbiome, which is disrupted following antibiotic treatment, may help overcome platinum resistance in patients with epithelial ovarian cancer. See related commentary by Hawkins and Nephew, p. 4511.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias Ováricas , Humanos , Femenino , Ratones , Animales , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/patología , Cisplatino/uso terapéutico , Estudios Retrospectivos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Ováricas/patología , Antibacterianos/farmacología
20.
Nat Rev Cancer ; 21(8): 526-536, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34103704

RESUMEN

Cellular heterogeneity and an immunosuppressive tumour microenvironment are independent yet synergistic drivers of tumour progression and underlie therapeutic resistance. Recent studies have highlighted the complex interaction between these cell-intrinsic and cell-extrinsic mechanisms. The reciprocal communication between cancer stem cells (CSCs) and infiltrating immune cell populations in the tumour microenvironment is a paradigm for these interactions. In this Perspective, we discuss the signalling programmes that simultaneously induce CSCs and reprogramme the immune response to facilitate tumour immune evasion, metastasis and recurrence. We further highlight biological factors that can impact the nature of CSC-immune cell communication. Finally, we discuss targeting opportunities for simultaneous regulation of the CSC niche and immunosurveillance.


Asunto(s)
Neoplasias/inmunología , Células Madre Neoplásicas/inmunología , Animales , Humanos , Neoplasias/patología , Procesos Neoplásicos , Escape del Tumor , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA