Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cancer Biol Ther ; 23(1): 1-8, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-36201632

RESUMEN

Stereotactic body radiotherapy (SBRT) demonstrates excellent local control in early stage lung cancer, however a quarter of patients develop recurrence or distant metastasis. Transforming growth factor-beta (TGF-ß) supports metastasis and treatment resistance, and angiotensin receptor blockade (ARB) indirectly suppresses TGF-ß signaling. This study investigates whether patients taking ARBs while undergoing SBRT for early stage lung cancer exhibited improved overall survival (OS) or recurrence free survival (RFS) compared to patients not taking ARBs. This was a single institution retrospective analysis of 272 patients treated with SBRT for early stage lung cancer between 2009 and 2018. Patient health data was abstracted from the electronic medical record. OS and RFS were assessed using Kaplan-Meier method. Log-rank test was used to compare unadjusted survival between groups. Univariable and multivariable Cox proportional hazard regression models were used to estimate hazard ratios (HRs). Of 247 patients analyzed, 24 (10%) patients took ARBs for the duration of radiotherapy. There was no difference in mean age, median tumor diameter, or median biologic effective dose between patients taking ARBs or not. Patients taking ARBs exhibited increased OS (ARB = 96.7 mo.; no ARB = 43.3 mo.; HR = 0.25 [95% CI: 0.10 to 0.62, P = .003]) and increased RFS (median RFS, ARB = 64.3 mo.; No ARB = 35.1 mo.; HR = 0.26 [95% CI: 0.10 to 0.63, P = .003]). These effects were not seen in patients taking angiotensin converting enzyme inhibitors (ACEIs) or statins. ARB use while undergoing SBRT for early stage lung cancer may increase OS and RFS, but ACEI use does not show the same effect.


Asunto(s)
Productos Biológicos , Carcinoma de Pulmón de Células no Pequeñas , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Neoplasias Pulmonares , Radiocirugia , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Productos Biológicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Radiocirugia/métodos , Receptores de Angiotensina/uso terapéutico , Estudios Retrospectivos , Factor de Crecimiento Transformador beta , Factores de Crecimiento Transformadores/uso terapéutico , Resultado del Tratamiento
2.
Lancet Oncol ; 23(9): 1189-1200, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35952709

RESUMEN

BACKGROUND: TGF-ß is an immunosuppressive cytokine that is upregulated in colorectal cancer. TGF-ß blockade improved response to chemoradiotherapy in preclinical models of colorectal adenocarcinoma. We aimed to test the hypothesis that adding the TGF-ß type I receptor kinase inhibitor galunisertib to neoadjuvant chemoradiotherapy would improve pathological complete response rates in patients with locally advanced rectal cancer. METHODS: This was an investigator-initiated, single-arm, phase 2 study done in two medical centres in Portland (OR, USA). Eligible patients had previously untreated, locally advanced, rectal adenocarcinoma, stage IIA-IIIC or IV as per the American Joint Committee on Cancer; Eastern Cooperative Oncology Group status 0-2; and were aged 18 years or older. Participants completed two 14-day courses of oral galunisertib 150 mg twice daily, before and during fluorouracil-based chemoradiotherapy (intravenous fluorouracil 225 mg/m2 over 24 h daily 7 days per week during radiotherapy or oral capecitabine 825 mg/m2 twice per day 5 days per week during radiotherapy; radiotherapy consisted of 50·4-54·0 Gy in 28-30 fractions). 5-9 weeks later, patients underwent response assessment. Patients with a complete response could opt for non-operative management and proceed to modified FOLFOX6 (intravenous leucovorin 400 mg/m2 on day 1, intravenous fluorouracil 400 mg/m2 on day 1 then 2400 mg/m2 over 46 h, and intravenous oxaliplatin 85 mg/m2 on day 1 delivered every 2 weeks for eight cycles) or CAPEOX (intravenous oxaliplatin 130 mg/m2 on day 1 and oral capecitabine 1000 mg/m2 twice daily for 14 days every 3 weeks for four cycles). Patients with less than complete response underwent surgical resection. The primary endpoint was complete response rate, which was a composite of pathological complete response in patients who proceeded to surgery, or clinical complete response maintained at 1 year after last therapy in patients with non-operative management. Safety was a coprimary endpoint. Both endpoints were assessed in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT02688712, and is active but not recruiting. FINDINGS: Between Oct 19, 2016, and Aug 31, 2020, 38 participants were enrolled. 25 (71%) of the 35 patients who completed chemoradiotherapy proceeded to total mesorectal excision surgery, five (20%) of whom had pathological complete responses. Ten (29%) patients had non-operative management, three (30%) of whom ultimately chose to have total mesorectal excision. Two (67%) of those three patients had pathological complete responses. Of the remaining seven patients in the non-operative management group, five (71%) had clinical complete responses at 1 year after their last modified FOLFOX6 infusion. In total, 12 (32% [one-sided 95% CI ≥19%]) of 38 patients had a complete response. Common grade 3 adverse events during treatment included diarrhoea in six (16%) of 38 patients, and haematological toxicity in seven (18%) patients. Two (5%) patients had grade 4 adverse events, one related to chemoradiotherapy-induced diarrhoea and dehydration, and the other an intraoperative ischaemic event. No treatment-related deaths occurred. INTERPRETATION: The addition of galunisertib to neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer improved the complete response rate to 32%, was well tolerated, and warrants further assessment in randomised trials. FUNDING: Eli Lilly via ExIST program, The Providence Foundation.


Asunto(s)
Adenocarcinoma , Neoplasias Primarias Secundarias , Neoplasias del Recto , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Capecitabina , Quimioradioterapia/efectos adversos , Diarrea/etiología , Fluorouracilo , Humanos , Terapia Neoadyuvante/efectos adversos , Estadificación de Neoplasias , Neoplasias Primarias Secundarias/patología , Oxaliplatino , Pirazoles , Quinolinas , Neoplasias del Recto/tratamiento farmacológico , Neoplasias del Recto/patología , Factor de Crecimiento Transformador beta
3.
Mol Carcinog ; 61(2): 165-172, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34644811

RESUMEN

Radiation has been utilized as a direct cytotoxic tumorcidal modality, however, the effect of radiation on tumor vasculature influences response to anticancer therapies. Although numerous reports have demonstrated vascular changes in irradiated tumors, the findings and implications are extensive and at times contradictory depending on the radiation dose, timing, and models used. In this review, we focus on the radiation-mediated effects on tumor vasculature with respect to doses used, timing postradiation, vasculogenesis, adhesion molecule expression, permeability, and pericyte coverage, including the latest findings.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Neovascularización Patológica/tratamiento farmacológico
4.
Oncoimmunology ; 10(1): 1900635, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33796412

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) has traditionally been thought of as an immunologically quiescent tumor type presumably because of a relatively low tumor mutational burden (TMB) and poor responses to checkpoint blockade therapy. However, many PDAC tumors exhibit T cell inflamed phenotypes. The presence of tertiary lymphoid structures (TLS) has recently been shown to be predictive of checkpoint blockade response in melanomas and sarcomas, and are prognostic for survival in PDAC. In order to more comprehensively understand tumor immunity in PDAC patients with TLS, we performed RNA-seq, single and multiplex IHC, flow cytometry and predictive genomic analysis on treatment naïve, PDAC surgical specimens. Forty-six percent of tumors contained distinct T and B cell aggregates reflective of "early-stage TLS" (ES-TLS), which correlated with longer overall and progression-free survival. These tumors had greater CD8+ T cell infiltration but were not defined by previously published TLS gene-expression signatures. ES-TLS+ tumors were enriched for IgG1 class-switched memory B cells and memory CD4+ T cells, suggesting durable immunological memory persisted in these patients. We also observed the presence of active germinal centers (mature-TLS) in 31% of tumors with lymphocyte clusters, whose patients had long-term survival (median 56 months). M-TLS-positive tumors had equivalent overall T cell infiltration to ES-TLS, but were enriched for activated CD4+ memory cells, naive B cells and NK cells. Finally, using a TCGA-PDAC dataset, ES-TLS+ tumors harbored a decreased TMB, but M-TLS with germinal centers expressed significantly more MHCI-restricted neoantigens as determined by an in silico neoantigen prediction method. Interestingly, M-TLS+ tumors also had evidence of increased rates of B cell somatic hypermutation, suggesting that germinal centers form in the presence of high-quality tumor neoantigens leading to increased humoral immunity that confers improved survival for PDAC patients. AbbreviationsTLS: tertiary lymphoid structures; GC: germinal center(s); PDAC: pancreatic ductal adenocarcinoma; RNA-seq: RNA sequencing; BCRseq: B cell receptor sequencing; HEV: high endothelial venule; PNAd: peripheral node addressin; TMB: tumor mutational burden; TCGA: the cancer genome atlas; PAAD: pancreatic adenocarcinoma; FFPE: formalin fixed paraffin embedded; TIME: tumor immune microenvironment.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Estructuras Linfoides Terciarias , Centro Germinal , Humanos , Inmunidad Humoral , Neoplasias Pancreáticas/genética , Supervivencia , Microambiente Tumoral
5.
Artículo en Inglés | MEDLINE | ID: mdl-32753776

RESUMEN

In many applications based on kinetic evaluation analysis and model fitting, quantitative mapping retrieved on data series from modalites such as MRI is completed on a voxel-by-voxel basis, where motion and low signal to noise ratio (SNR) would considerably degenerate the reliability of estimations. The coherence of image series in space and time can be used as prior knowledge to mitigate this occurrence. In this study, spatial and temporal higher-order total variations (HOTVs) are applied on a data series of MRI signal (e.g. dynamic contrast-enhanced (DCE) MRI and intravoxel incoherent motion (IVIM) MRI) to exploit the coherence of signal in space and time to minimize the variabilities caused by motion as well as improving quality of images with low SNR while retaining the physical details of original data properly. Simultaneously applying spatial and temporal HOTVs on images is non-trivial in implementation since it is a non-smooth optimization problem with multiple regularizers. Therefore, we use the proximal gradient method as well as a primal-dual split proximal mechanism to address the problem properly. In addition to increase the reliability of quantitative parametric map estimations, this preprocessing procedure can be included into many existing map estimation algorithms and pipelines effortlessly. We demonstrate our method on the parametric maps estimation for DCE MRI and IVIM MRI.

6.
Oral Oncol ; 109: 104770, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32599498

RESUMEN

BACKGROUND: In this study we determine the survival in patients with HPV-positive oropharyngeal carcinoma treated with transoral robotic surgery (TORS), neck dissection and risk-adapted adjuvant therapy. METHODS: We retrospectively identified 122 patients with HPV-positive oropharyngeal carcinoma treated with TORS and neck dissection between 2011 and 2018. Survival probability was calculated. We determined the effect of the type of neck dissection performed (modified radical neck dissection-MRND vs. selective neck dissection - SND), extranodal extension (ENE), margin status, and presence of ≥ 5 metastatic nodes on survival. RESULTS: Our patient population had a five-year overall survival of 91.0% (95% C.I. 85-97%). The five-year probability of recurrence or cancer-associated death was 0.0977 (95% C.I. 0.0927-0.1027). The five-year probability of cancer-associated death was 0.0528 (95% C.I. 0.048-0.0570). All patients who died of their disease had distant metastasis. Our PEG dependence rate was 0%. Patients with ENE and positive margins who underwent adjuvant chemoradiation did not have worse survival. Presence of ≥ 5 metastatic nodes portended worse survival after controlling for age, positive ENE and margins. Low yield (<18 nodes) on neck dissection worsened DFS on multivariable analysis. Furthermore, patients who underwent SND did not have worse OS than those who underwent MRND. CONCLUSION: Our study demonstrates that surgery could be simplified by performing TORS with SND rather than MRND. The one true poor prognostic factor in HPV-positive oropharyngeal carcinoma patients who undergo surgery is high nodal burden. Patients with high nodal burden are much more likely to die from their disease.

7.
Semin Radiat Oncol ; 30(2): 158-172, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32381295

RESUMEN

Immunotherapies are rapidly entering the clinic as approved treatments for diverse cancer pathologies. Radiation therapy is an integral partner in cancer therapy, commonly as part of complicated multimodality approaches that optimize patient outcomes. Preclinical studies have demonstrated that the success of radiation therapy in tumor control is due in part to immune mechanisms, and that outcomes following radiation therapy can be improved through combination with a range of immunotherapies. However, preclinical models of cancer are very different from patient tumors, and the way these preclinical tumors are treated is often very different from standard of care treatment of patients. This review examines the preclinical and clinical data for the role of the immune system in radiation therapy outcomes, and how to integrate preclinical findings into clinical trials, using ongoing studies as examples.


Asunto(s)
Ensayos Clínicos como Asunto , Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/radioterapia , Proyectos de Investigación , Animales , Terapia Combinada , Modelos Animales de Enfermedad , Humanos
8.
Nat Commun ; 11(1): 1749, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32273499

RESUMEN

Transforming growth factor beta (TGFß) is a multipotent immunosuppressive cytokine. TGFß excludes immune cells from tumors, and TGFß inhibition improves the efficacy of cytotoxic and immune therapies. Using preclinical colorectal cancer models in cell type-conditional TGFß receptor I (ALK5) knockout mice, we interrogate this mechanism. Tumor growth delay and radiation response are unchanged in animals with Treg or macrophage-specific ALK5 deletion. However, CD8αCre-ALK5flox/flox (ALK5ΔCD8) mice reject tumors in high proportions, dependent on CD8+ T cells. ALK5ΔCD8 mice have more tumor-infiltrating effector CD8+ T cells, with more cytotoxic capacity. ALK5-deficient CD8+ T cells exhibit increased CXCR3 expression and enhanced migration towards CXCL10. TGFß reduces CXCR3 expression, and increases binding of Smad2 to the CXCR3 promoter. In vivo CXCR3 blockade partially abrogates the survival advantage of an ALK5ΔCD8 host. These data demonstrate a mechanism of TGFß immunosuppression through inhibition of CXCR3 in CD8+ T cells, thereby limiting their trafficking into tumors.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Neoplasias/genética , Receptores CXCR3/genética , Factor de Crecimiento Transformador beta/farmacología , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/metabolismo , Neoplasias/patología , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Receptores CXCR3/metabolismo , Proteína Smad2/metabolismo
9.
PLoS One ; 14(2): e0211117, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30726287

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a fibrotic stroma with a poor lymphocyte infiltrate, in part driven by cancer-associated fibroblasts (CAFs). CAFs, which express fibroblast activation protein (FAP), contribute to immune escape via exclusion of anti-tumor CD8+ T cells from cancer cells, upregulation of immune checkpoint ligand expression, immunosuppressive cytokine production, and polarization of tumor infiltrating inflammatory cells. FAP is a post-proline peptidase selectively expressed during tissue remodeling and repair, such as with wound healing, and in the tumor microenvironment by cancer-associated fibroblasts. We targeted FAP function using a novel small molecule inhibitor, UAMC-1110, and mice with germline knockout of FAP and concomitant knock-in of E. coli beta-galactosidase. We depleted CAFs by adoptive transfer of anti-ßgal T cells into the FAP knockout animals. Established syngeneic pancreatic tumors in immune competent mice were targeted with these 3 strategies, followed by focal radiotherapy to the tumor. FAP loss was associated with improved antigen-specific tumor T cell infiltrate and enhanced collagen deposition. However, FAP targeting alone or with tumor-directed radiation did not improve survival even when combined with anti-PD1 therapy. Targeting of CAFs alone or in combination with radiation did not improve survival. We conclude that targeting FAP and CAFs in combination with radiation is capable of enhancing anti-tumor T cell infiltrate and function, but does not result in sufficient tumor clearance to extend survival.


Asunto(s)
Anticuerpos/metabolismo , Carcinoma Ductal Pancreático/terapia , Gelatinasas/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Neoplasias Pancreáticas/terapia , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Linfocitos T/trasplante , Traslado Adoptivo , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Quimioradioterapia , Terapia Combinada , Endopeptidasas , Gelatinasas/genética , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Humanos , Proteínas de la Membrana/genética , Ratones , Neoplasias Pancreáticas/metabolismo , Serina Endopeptidasas/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Linfocitos T/inmunología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto , beta-Galactosidasa/inmunología
10.
Adv Radiat Oncol ; 3(4): 494-505, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30370348

RESUMEN

PURPOSE: The purpose of this article is to assemble, review, and provide a synopsis of the historical and current literature regarding optimal sequencing of radiation (RT) and immunotherapy combination treatments. MATERIALS AND METHODS: A review of the literature was performed using PubMed with the query "radiation" and "Immunotherapy", "PD1", "PDL1", "CTLA4", "OX40", "checkpoint", "vaccine", "macrophage", "STING", and "TGFbeta". Studies that included sequencing of therapy were evaluated and the studies were included at the authors discretion. RESULTS: A paucity of primary literature exists examining the best order of radiation and immunotherapy, most of which was performed in the pre-clinical setting. The observations are that optimal sequencing of various radiation plus immune therapy combinations is dependent on the mechanism(s) of activation by the combination treatment. Immunosuppressive molecules tend to be better inhibited prior to RT while engagement of costimulatory genes is better activated concomitantly with RT. CONCLUSIONS: These data should compel more basic research into both the direct investigation of sequencing efficacy and studies on the mechanisms of immune mediated cell death potentiated by radio-therapy.

12.
PLoS One ; 12(11): e0187532, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29135982

RESUMEN

Human papilloma virus positive (HPV+) tumors represent a large proportion of anal, vulvar, vaginal, cervical and head and neck squamous carcinomas (HNSCC) and late stage invasive disease is thought to originate from a premalignant state. Cyclic dinucleotides that activate STimulator of INterferon Genes (STING) have been shown to cause rapid regression of a range of advanced tumors. We aimed to investigate STING ligands as a novel treatment for papilloma. We tested therapies in a spontaneous mouse model of papilloma of the face and anogenital region that histologically resembles human HPV-associated papilloma. We demonstrate that STING ligands cause rapid regression of papilloma, associated with T cell infiltration, and are significantly more effective than Imiquimod, a current immunotherapy for papilloma. In humans, we show that STING is expressed in the basal layer of normal skin and lost during keratinocyte differentiation. We found STING was expressed in all HPV-associated cervical and anal dysplasia and was strongly expressed in the cancer cells of HPV+ HNSCC but not in HPV-unrelated HNSCC. We found no strong association between STING expression and progressive disease in non-HPV oral dysplasia and oral pre-malignancies that are not HPV-related. These data demonstrate that STING is expressed in basal cells of the skin and is retained in HPV+ pre-malignancies and advanced cancers, but not in HPV-unrelated HNSCC. However, using a murine HNSCC model that does not express STING, we demonstrate that STING ligands are an effective therapy regardless of expression of STING by the cancer cells.


Asunto(s)
Alphapapillomavirus/aislamiento & purificación , Proteínas de la Membrana/metabolismo , Neoplasias/virología , Lesiones Precancerosas/virología , Animales , Femenino , Humanos , Ligandos , Masculino , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/metabolismo
13.
Oncotarget ; 8(6): 9155-9173, 2017 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-27893434

RESUMEN

BACKGROUND: Cancer patients often report behavioral and cognitive changes following cancer treatment. These effects can be seen in patients who have not yet received treatment or have received only peripheral (non-brain) irradiation. Novel treatments combining radiotherapy (RT) and immunotherapy (IT) demonstrate remarkable efficacy with respect to tumor outcomes by enhancing the proinflammatory environment in the tumor. However, a proinflammatory environment in the brain mediates cognitive impairments in other neurological disorders and may affect brain function in cancer patients receiving these novel treatments. Currently, gaps exist as to whether these treatments impact the brain in individuals with or without tumors and with regard to the underlying mechanisms. RESULTS: Combined treatment with precision RT and checkpoint inhibitor IT achieved control of tumor growth. However, BALB/c mice receiving combined treatment demonstrated changes in measures of anxiety levels, regardless of tumor status. C57BL/6J mice with tumors demonstrated increased anxiety, except following combined treatment. Object recognition memory was impaired in C57BL/6J mice without tumors following combined treatment. All mice with tumors showed impaired object recognition, except those treated with RT alone. Mice with tumors demonstrated impaired amygdala-dependent cued fear memory, while maintaining hippocampus-dependent context fear memory. These behavioral alterations and cognitive impairments were accompanied by increased microglial activation in mice receiving immunotherapy alone or combined with RT. Finally, based on tumor status, there were significant changes in proinflammatory cytokines (IFN-γ, IL-6, IL-5, IL-2, IL-10) and a growth factor (FGF-basic). MATERIALS AND METHODS: Here we test the hypothesis that IT combined with peripheral RT have detrimental behavioral and cognitive effects as a result of an enhanced proinflammatory environment in the brain. BALB/c mice with or without injected hind flank CT26 colorectal carcinoma or C57BL/6J mice with or without Lewis Lung carcinoma were used for all experiments. Checkpoint inhibitor IT, using an anti-CTLA-4 antibody, and precision CT-guided peripheral RT alone and combined were used to closely model clinical treatment. We assessed behavioral and cognitive performance and investigated the immune environment using immunohistochemistry and multiplex assays to analyze proinflammatory mediators. CONCLUSIONS: Although combined treatment achieved tumor growth control, it affected the brain and induced changes in measures of anxiety, cognitive impairments, and neuroinflammation.


Asunto(s)
Antineoplásicos Inmunológicos/toxicidad , Conducta Animal , Encéfalo , Carcinoma Pulmonar de Lewis/terapia , Trastornos del Conocimiento/etiología , Cognición , Neoplasias Colorrectales/terapia , Encefalitis/etiología , Traumatismos por Radiación/etiología , Radioinmunoterapia/efectos adversos , Animales , Ansiedad/etiología , Ansiedad/psicología , Aprendizaje por Asociación , Conducta Animal/efectos de los fármacos , Conducta Animal/efectos de la radiación , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/fisiopatología , Encéfalo/efectos de la radiación , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Cognición/efectos de los fármacos , Cognición/efectos de la radiación , Trastornos del Conocimiento/inmunología , Trastornos del Conocimiento/fisiopatología , Trastornos del Conocimiento/psicología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Citocinas/inmunología , Citocinas/metabolismo , Depresión/etiología , Depresión/psicología , Encefalitis/inmunología , Encefalitis/fisiopatología , Encefalitis/psicología , Conducta Exploratoria , Femenino , Memoria , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Actividad Motora , Comportamiento de Nidificación , Traumatismos por Radiación/inmunología , Traumatismos por Radiación/fisiopatología , Traumatismos por Radiación/psicología , Dosificación Radioterapéutica , Reconocimiento en Psicología , Factores de Tiempo
14.
Hepatol Res ; 47(7): 702-714, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27501850

RESUMEN

AIM: Small, solitary hepatocellular carcinoma is curable with stereotactic radiation or other methods of tumor ablation, however, regional and systemic tumor recurrence occurs in over 70% of patients. Here we describe the ability of immunoradiotherapy to induce an antitumor immune response and delay the growth of tumors in immunocompetent mice. METHODS: A syngeneic hepatocellular carcinoma cell line (Hep-55.1c) was injected directly into the livers of C57BL/6 mice using ultrasound guidance, then tumors were treated with stereotactic radiation using a Small Animal Radiation Research Platform with computed tomography guidance. RESULTS: Delivery of three doses of 250 µg anti-programmed cell death protein-1 (αPD-1) antibody concurrently with 30 Gy stereotactic body radiation therapy in three fractions reduced the growth rate of tumors and improved survival (P < 0.05). Combined treatment was associated with increased CD8+ cytotoxic T cells in the tumor; depletion of CD8 T cells eliminated the efficacy of combined treatment. Combined treatment also induced expression of programmed cell death-1 ligand expression on tumor-infiltrating macrophages, and the tumors grew rapidly after αPD-1 treatment was discontinued. CONCLUSIONS: Tumor response to stereotactic radiation can be augmented by concurrent treatment with αPD-1. The efficacy of this combination therapy was transient, however, and treatment induced markers of adaptive immune resistance. These data are promising, but also indicate that mechanisms of immune resistance will need to be durably overcome for this combination to generate lasting immunity to protect against tumor recurrence.

15.
PLoS One ; 11(6): e0157164, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27281029

RESUMEN

The anecdotal reports of promising results seen with immunotherapy and radiation in advanced malignancies have prompted several trials combining immunotherapy and radiation. However, the ideal timing of immunotherapy with radiation has not been clarified. Tumor bearing mice were treated with 20Gy radiation delivered only to the tumor combined with either anti-CTLA4 antibody or anti-OX40 agonist antibody. Immunotherapy was delivered at a single timepoint around radiation. Surprisingly, the optimal timing of these therapies varied. Anti-CTLA4 was most effective when given prior to radiation therapy, in part due to regulatory T cell depletion. Administration of anti-OX40 agonist antibody was optimal when delivered one day following radiation during the post-radiation window of increased antigen presentation. Combination treatment of anti-CTLA4, radiation, and anti-OX40 using the ideal timing in a transplanted spontaneous mammary tumor model demonstrated tumor cures. These data demonstrate that the combination of immunotherapy and radiation results in improved therapeutic efficacy, and that the ideal timing of administration with radiation is dependent on the mechanism of action of the immunotherapy utilized.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/inmunología , Neoplasias Colorrectales/terapia , Inmunoterapia , Neoplasias Mamarias Animales/terapia , Receptores OX40/inmunología , Linfocitos T Reguladores/inmunología , Animales , Presentación de Antígeno , Antígeno CTLA-4/metabolismo , Neoplasias Colorrectales/inmunología , Terapia Combinada , Fraccionamiento de la Dosis de Radiación , Femenino , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Oncoimmunology ; 4(3): e955696, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25949887

RESUMEN

The tumor immune environment has been linked to prognosis in patients with a range of malignancies. Recently, we demonstrated in pre-clinical models that modifying the tumor immune environment using a small-molecule inhibitor of TGFb significantly improved outcome to subsequent radiation therapy. These data suggest that this and other immunotherapies may be used to remodel the tumor before conventional cancer therapies to improve outcomes.

18.
Cancer Immunol Res ; 2(10): 1011-22, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25047233

RESUMEN

The immune infiltrate in colorectal cancer has been correlated with outcome, such that individuals with higher infiltrations of T cells have increased survival independent of the disease stage. For patients with lower immune infiltrates, overall survival is limited. Because the patients with colorectal cancer studied have received conventional cancer therapies, these data may indicate that the pretreatment tumor environment increases the efficacy of treatments such as chemotherapy, surgery, and radiotherapy. This study was designed to test the hypothesis that an improved immune environment in the tumor at the time of treatment will increase the efficacy of radiotherapy. We demonstrate that inhibition of TGFß using the orally available small-molecule inhibitor SM16 improved the immune environment of tumors in mice and significantly improved the efficacy of subsequent radiotherapy. This effect was not due to changes in radiosensitivity, epithelial-mesenchymal transition, or changes in vascular function in the tumor; rather, this effect was dependent on adaptive immunity and resulted in long-term protective immunity in cured mice. These data demonstrate that immunotherapy is an option to improve the immune status of patients with poor tumor infiltrates and that pretreatment improves the efficacy of radiotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Compuestos de Azabiciclo/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Inmunidad Adaptativa/efectos de los fármacos , Inmunidad Adaptativa/inmunología , Animales , Quimioterapia Adyuvante/métodos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/radioterapia , Evaluación Preclínica de Medicamentos/métodos , Femenino , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/radioterapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Neoadyuvante/métodos , Trasplante de Neoplasias , Tolerancia a Radiación/efectos de los fármacos , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...