Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Blood Cancer ; 70(6): e30324, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37017066

RESUMEN

BACKGROUND: Although tumor genomic profiling has aided the advancement of targeted genetic therapy, its clinical integration remains a challenge in pediatric cancers due to lower mutation frequency and less available targeted drugs. There have been multiple novel studies examining molecular sequencing in pediatrics; however, many of these studies primarily utilized large-scale, genome-wide screening applications that limit applicable use due to the availability of testing. This study examined the institutional use of a targeted, clinically available approach to tumor genomic sequencing. METHODS: A retrospective chart review was performed on pediatric patients with solid tumors who were managed at Roswell Park Comprehensive Cancer Center and underwent molecular testing of their tumor biopsy via OmniSeq from August 2016 to July 2021. Results were reviewed for mutations considered to be "actionable" by targeted therapy. Patients with actionable mutations were further examined to evaluate treatment course, receival of targeted therapy, and clinical outcomes. RESULTS: We identified 64 pediatric patients consisting of 20 (31%) with CNS tumors and 44 (69%) with non-CNS tumors, ranging in age from 9 months to 21 years. Thirty-five total actionable mutations were identified amongst 27 patients (42%). Of these 27, 12 patients (44%) received at least 1 targeted drug against a respective actionable mutation, of which 6 patients (50%) achieved clinical benefit to therapy, including 1 complete response. CONCLUSIONS: The use of a clinically focused and readily available targeted molecular sequencing panel identified actionable mutations at a comparable rate to the large-scale, less readily available sequencing panels utilized in other studies. Half of our patients who received targeted therapy achieved a complete response or clinical benefit from therapy. Although targeted therapy has a role in pediatric cancer treatment, many newer drugs require further research on their safety and efficacy.


Asunto(s)
Neoplasias , Medicina de Precisión , Humanos , Niño , Estudios Retrospectivos , Medicina de Precisión/métodos , Neoplasias/tratamiento farmacológico , Mutación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Genómica/métodos , Biomarcadores de Tumor/genética , Terapia Molecular Dirigida/métodos
2.
Front Immunol ; 13: 1023023, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36330525

RESUMEN

Background: Arginase-1-producing cells inhibit T cell-mediated anti-tumor responses by reducing L-arginine levels in the tumor microenvironment. T cell-facilitated elimination of arginase-1-expressing cells could potentially restore L-arginine levels and improve anti-tumor responses. The activation of arginase-1-specific T cells may convert the immunosuppressive tumor microenvironment and induce or strengthen local Th1 inflammation. In the current clinical study, we examined the safety and immunogenicity of arginase-1-based peptide vaccination. Methods: In this clinical phase I trial, ten patients with treatment-refractory progressive solid tumors were treated. The patients received an arginase-1 peptide vaccine comprising three 20-mer peptides from the ARG1 immunological "hot spot" region in combination with the adjuvant Montanide ISA-51. The vaccines were administered subcutaneously every third week (maximum 16 vaccines). The primary endpoint was to evaluate safety assessed by Common Terminology Criteria for Adverse Events 4.0 and laboratory monitoring. Vaccine-specific immune responses were evaluated using enzyme-linked immune absorbent spot assays and intracellular cytokine staining on peripheral blood mononuclear cells. Clinical responses were evaluated using Response Evaluation Criteria in Solid Tumors 1.1. Results: The vaccination was feasible, and no vaccine-related grade 3-4 adverse events were registered. Nine (90%) of ten patients exhibited peptide-specific immune responses in peripheral blood mononuclear cells. Six (86%) of the seven evaluable patients developed a reactive T cell response against at least one of the ARG1 peptides during treatment. A phenotypic classification revealed that arginase-1 vaccine-specific T cells were both CD4+ T cells and CD8+ T cells. Two (20%) of ten patients obtained stable disease for respectively four- and seven months on vaccination treatment. Conclusion: The peptide vaccine against arginase-1 was safe. Nine (90%) of ten patients had measurable peptide-specific responses in the periphery blood, and two (20%) of ten patients attained stable disease on protocol treatment. Clinical trial registration: https://clinicaltrials.gov/ct2/show/NCT03689192, identifier NCT03689192.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Humanos , Vacunas contra el Cáncer/efectos adversos , Vacunas de Subunidad/efectos adversos , Arginasa , Leucocitos Mononucleares , Neoplasias/tratamiento farmacológico , Péptidos/uso terapéutico , Arginina , Microambiente Tumoral
3.
Biomolecules ; 12(11)2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36358931

RESUMEN

A potential target of precision nutrition in cancer therapeutics is the micronutrient selenium (Se). Se is metabolized and incorporated as the amino acid selenocysteine (Sec) into 25 human selenoproteins, including glutathione peroxidases (GPXs) and thioredoxin reductases (TXNRDs), among others. Both the processes of Se and Sec metabolism for the production of selenoproteins and the action of selenoproteins are utilized by cancer cells from solid tumors as a protective mechanism against oxidative damage and to resist ferroptosis, an iron-dependent cell death mechanism. Protection against ferroptosis in cancer cells requires sustained production of the selenoprotein GPX4, which involves increasing the uptake of Se, potentially activating Se metabolic pathways such as the trans-selenation pathway and the TXNRD1-dependent decomposition of inorganic selenocompounds to sustain GPX4 synthesis. Additionally, endoplasmic reticulum-resident selenoproteins also affect apoptotic responses in the presence of selenocompounds. Selenoproteins may also help cancer cells adapting against increased oxidative damage and the challenges of a modified nutrient metabolism that result from the Warburg switch. Finally, cancer cells may also rewire the selenoprotein hierarchy and use Se-related machinery to prioritize selenoproteins that are essential to the adaptations against ferroptosis and oxidative damage. In this review, we discuss both the evidence and the gaps in knowledge on how cancer cells from solid tumors use Se, Sec, selenoproteins, and the Se-related machinery to promote their survival particularly via resistance to ferroptosis.


Asunto(s)
Ferroptosis , Neoplasias , Selenio , Humanos , Selenocisteína/metabolismo , Selenio/metabolismo , Selenoproteínas/metabolismo , Neoplasias/patología
4.
Expert Opin Drug Deliv ; 19(8): 899-912, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35830722

RESUMEN

INTRODUCTION: Many small molecules and biologic therapeutics have been developed for solid tumor therapy. However, the unique physiology of tumors makes the actual delivery of these drugs into the tumor mass inefficient. Such delivery requires transport from blood vessels, across the vasculature and into and through interstitial space within a tumor. This transportation is dependent on the physiochemical properties of the therapeutic agent and the biological properties of the tumor. It was hoped the application of nanoscale drug carrier systems would solve this problem. However, issues with poor tumor accumulation and limited drug release have impeded clinical impact. In response, these carrier systems have been redesigned to be paired with targetable external mechanical stimuli which can trigger much enhanced drug release and deposition. AREAS COVERED: The pre-clinical and clinical progress of thermolabile drug carrier systems and the modalities used to trigger the release of their cargo are assessed. EXPERT OPINION: Combined application of mild hyperthermia and heat-responsive liposomal drug carriers has great potential utility. Clinical trials continue to progress this approach and serve to refine the technologies, dosing regimens and exposure parameters that will provide optimal patient benefit.


Asunto(s)
Antineoplásicos , Hipertermia Inducida , Neoplasias , Doxorrubicina , Portadores de Fármacos/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Liposomas/química , Neoplasias/tratamiento farmacológico
5.
Zhonghua Zhong Liu Za Zhi ; 44(1): 112-119, 2022 Jan 23.
Artículo en Chino | MEDLINE | ID: mdl-35073657

RESUMEN

Objective: To investigate the feasibility, safety and efficacy of intrathecal pemetrexed (IP) treated for patients with leptomeningeal metastases (LM) from solid tumors. Methods: Forty-seven patients receiving pemetrexed intrathecal chemotherapy in the First Hospital of Jilin University from 2017 to 2018 were selected. The study of pemetrexed intrathecal chemotherapy adopted the classical dose-climbing model and included 13 patients with meningeal metastasis of non-small cell lung cancer who had relapsed and refractory after multiple previous treatments including intrathecal chemotherapy. Based on the dose climbing study, 34 patients with meningeal metastasis of solid tumor who did not receive intrathecal chemotherapy were enrolled in a clinical study using pemetrexed as the first-line intrathecal chemotherapy combined with radiotherapy. Kaplan-Meier method and Log rank test were used for survival analysis, and Cox regression model was used for influencing factor analysis. Results: The dose climbing study showed that the maximum tolerated dose of pemetrexed intrathecal chemotherapy was 10 mg per single dose, and the recommended dosing regimen was 10 mg once or twice a week. The incidence of adverse reactions was 10 cases, including hematological adverse reactions (7 cases), transaminase elevation (2 cases), nerve root reactions (5 cases), fatigue and weight loss (1 case). The incidence of serious adverse reactions was 4, including grade 4-5 poor hematology (2 cases), grade 4 nerve root irritation (2 cases), and grade 4 elevated aminotransferase (1 case). In the dose climbing study, 4 patients were effectively treated and 7 were disease controlled. The survival time was ranged from 0.3 to 14.0 months and a median survival time was 3.8 months. The clinical study of pemetrexed intrathecal chemotherapy combined with radiotherapy showed that the treatment mode of 10 mg pemetrexed intrathecal chemotherapy once a week combined with synchronous involved area radiotherapy 40 Gy/4 weeks had a high safety and reactivity. The incidence of major adverse reactions was 52.9% (18/34), including hematologic adverse reactions (13 cases), transaminase elevation (10 cases), and nerve root reactions (4 cases). In study 2, the response rate was 67.6% (23/34), the disease control rate was 73.5% (25/34), the overall survival time was ranged from 0.3 to 16.6 months, the median survival time was 5.5 months, and the 1-year survival rate was 21.6%. Clinical response, improvement of neurological dysfunction, completion of concurrent therapy and subsequent systemic therapy were associated with the overall survival (all P<0.05). Conclusions: Pemetrexed is suitable for the intrathecal chemotherapy with a high safety and efficacy. The recommended administration regimen was IP at 10 mg on the schedule of once or twice per week. Hematological toxicity is the main factor affecting the implementation of IP. Vitamin supplement can effectively control the occurrence of hematological toxicity.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinomatosis Meníngea , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Carcinomatosis Meníngea/tratamiento farmacológico , Pemetrexed , Resultado del Tratamiento
6.
Chinese Journal of Oncology ; (12): 112-119, 2022.
Artículo en Chino | WPRIM | ID: wpr-935190

RESUMEN

Objective: To investigate the feasibility, safety and efficacy of intrathecal pemetrexed (IP) treated for patients with leptomeningeal metastases (LM) from solid tumors. Methods: Forty-seven patients receiving pemetrexed intrathecal chemotherapy in the First Hospital of Jilin University from 2017 to 2018 were selected. The study of pemetrexed intrathecal chemotherapy adopted the classical dose-climbing model and included 13 patients with meningeal metastasis of non-small cell lung cancer who had relapsed and refractory after multiple previous treatments including intrathecal chemotherapy. Based on the dose climbing study, 34 patients with meningeal metastasis of solid tumor who did not receive intrathecal chemotherapy were enrolled in a clinical study using pemetrexed as the first-line intrathecal chemotherapy combined with radiotherapy. Kaplan-Meier method and Log rank test were used for survival analysis, and Cox regression model was used for influencing factor analysis. Results: The dose climbing study showed that the maximum tolerated dose of pemetrexed intrathecal chemotherapy was 10 mg per single dose, and the recommended dosing regimen was 10 mg once or twice a week. The incidence of adverse reactions was 10 cases, including hematological adverse reactions (7 cases), transaminase elevation (2 cases), nerve root reactions (5 cases), fatigue and weight loss (1 case). The incidence of serious adverse reactions was 4, including grade 4-5 poor hematology (2 cases), grade 4 nerve root irritation (2 cases), and grade 4 elevated aminotransferase (1 case). In the dose climbing study, 4 patients were effectively treated and 7 were disease controlled. The survival time was ranged from 0.3 to 14.0 months and a median survival time was 3.8 months. The clinical study of pemetrexed intrathecal chemotherapy combined with radiotherapy showed that the treatment mode of 10 mg pemetrexed intrathecal chemotherapy once a week combined with synchronous involved area radiotherapy 40 Gy/4 weeks had a high safety and reactivity. The incidence of major adverse reactions was 52.9% (18/34), including hematologic adverse reactions (13 cases), transaminase elevation (10 cases), and nerve root reactions (4 cases). In study 2, the response rate was 67.6% (23/34), the disease control rate was 73.5% (25/34), the overall survival time was ranged from 0.3 to 16.6 months, the median survival time was 5.5 months, and the 1-year survival rate was 21.6%. Clinical response, improvement of neurological dysfunction, completion of concurrent therapy and subsequent systemic therapy were associated with the overall survival (all P<0.05). Conclusions: Pemetrexed is suitable for the intrathecal chemotherapy with a high safety and efficacy. The recommended administration regimen was IP at 10 mg on the schedule of once or twice per week. Hematological toxicity is the main factor affecting the implementation of IP. Vitamin supplement can effectively control the occurrence of hematological toxicity.


Asunto(s)
Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Carcinomatosis Meníngea/tratamiento farmacológico , Pemetrexed , Resultado del Tratamiento
7.
Nano Lett ; 21(24): 10267-10278, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34878286

RESUMEN

Tumor theranostics hold great potential for personalized medicine in the future, and transcatheter arterial embolization (TAE) is an important clinical treatment for unresectable or hypervascular tumors. In order to break the limitation, simplify the procedure of TAE, and achieve ideal combinatorial theranostic capability, here, a kind of triblock-polypeptide-coated perfluoropentane-loaded mesoporous Fe3O4 nanocomposites (PFP-m-Fe3O4@PGTTCs) were prepared for non-interventional target-embolization, magnetic hyperthermia, and multimodal imaging combination theranostics of solid tumors. The results of systematic animal experiments by H22-tumor-bearing mice and VX2-tumor-bearing rabbits in vivo indicated that PFP-m-Fe3O4@PGTTC-6.3 has specific tumor accumulation and embolization effects. The tumors' growth has been inhibited and the tumors disappeared 4 weeks and ≤15 days post-injection with embolization and magnetic hyperthermia combination therapy, respectively. The results also showed an excellent effect of magnetic resonance/ultrasound/SPECT multimodal imaging. This pH-responsive non-interventional embolization combinatorial theranostics system provides a novel embolization and multifunctional theranostic candidate for solid tumors.


Asunto(s)
Embolización Terapéutica , Hipertermia Inducida , Nanopartículas , Neoplasias , Animales , Hipertermia Inducida/métodos , Ratones , Imagen Multimodal/métodos , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Péptidos , Medicina de Precisión , Conejos
8.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34948200

RESUMEN

Combination therapy is becoming imperative for the treatment of many cancers, as it provides a higher chance of avoiding drug resistance and tumor recurrence. Among the resistance-conferring factors, the tumor microenvironment plays a major role, and therefore, represents a viable target for adjuvant therapeutic agents. Thus, hypoxia and extracellular acidosis are known to select for the most aggressive and resilient phenotypes and build poorly responsive regions of the tumor mass. Carbonic anhydrase (CA, EC 4.2.1.1) IX isoform is a surficial zinc metalloenzyme that is proven to play a central role in regulating intra and extracellular pH, as well as modulating invasion and metastasis processes. With its strong association and distribution in various tumor tissues and well-known druggability, this protein holds great promise as a target to pharmacologically interfere with the tumor microenvironment by using drug combination regimens. In the present review, we summarized recent publications revealing the potential of CA IX inhibitors to intensify cancer chemotherapy and overcome drug resistance in preclinical settings.


Asunto(s)
Anhidrasa Carbónica IX/antagonistas & inhibidores , Inhibidores de Anhidrasa Carbónica/farmacología , Neoplasias/tratamiento farmacológico , Animales , Hipoxia de la Célula/efectos de los fármacos , Terapia Combinada , Humanos , Neoplasias/metabolismo , Microambiente Tumoral/efectos de los fármacos
9.
Asian Pac J Cancer Prev ; 22(10): 3211-3217, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34710997

RESUMEN

PURPOSE: Oral mucositis is a painful condition that occurs in patients who undergo chemotherapy. Due to the worsening of oral mucositis, the patient may progress to a worse clinical condition and interrupt antineoplastic treatment. There is little literature on low-power laser therapy in chemotherapy for other solid tumors. The purpose of this study was to investigate whether low-level laser therapy (LLLT) applied before chemotherapy could prevent oral mucositis in patients with solid tumors. METHODS: Laser therapy was applied at a frequency of 630nm, with a dose of 2J / cm2, for the prevention of oral mucositis induced by chemotherapy specifically for non-hematological tumors. Epidemiological data, total neutrophils, general side effects, development of oral mucositis and degree, and the performance of low-power laser therapy to prevent oral mucositis were collected. The involvement of oxidative stress was evaluated by the enzyme superoxide dismutase (SOD) through blood samples, before and after chemotherapy treatments. RESULTS: LLLT in the proposed protocol is efficient in reducing the development of oral mucositis (only at grade I/II) in patients under chemotherapy and able to reduce the severity of oral mucosal lesions, in patients who developed mucositis after the use of the laser for prevention. All individuals who underwent LLLT protocol did not show a significant reduction of SOD activity after the last chemotherapy cycle. CONCLUSIONS: The prophylactic laser therapy protocol proposed by the study, defined at a frequency of 630nm, a dose of 2J / cm2, demonstrated the ability to decrease the occurrence of oral mucositis in patients undergoing chemotherapy protocols to solid tumors. This effect could be related to preserved SOD activity, as it was observed that oral mucositis is related to leukopenia and reduced SOD activity and LLLT protocol prevented the decrease of SOD activity.


Asunto(s)
Terapia por Luz de Baja Intensidad/métodos , Neoplasias/tratamiento farmacológico , Estomatitis/prevención & control , Superóxido Dismutasa/sangre , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carboplatino/efectos adversos , Carboplatino/uso terapéutico , Cisplatino/efectos adversos , Cisplatino/uso terapéutico , Ciclofosfamida/efectos adversos , Ciclofosfamida/uso terapéutico , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Femenino , Fluorouracilo/efectos adversos , Fluorouracilo/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/enzimología , Neoplasias/patología , Estrés Oxidativo , Paclitaxel/efectos adversos , Paclitaxel/uso terapéutico , Estomatitis/inducido químicamente , Estomatitis/enzimología
10.
Cell Host Microbe ; 29(10): 1573-1588.e7, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34453895

RESUMEN

Despite overall success, T cell checkpoint inhibitors for cancer treatment are still only efficient in a minority of patients. Recently, intestinal microbiota was found to critically modulate anti-cancer immunity and therapy response. Here, we identify Clostridiales members of the gut microbiota associated with a lower tumor burden in mouse models of colorectal cancer (CRC). Interestingly, these commensal species are also significantly reduced in CRC patients compared with healthy controls. Oral application of a mix of four Clostridiales strains (CC4) in mice prevented and even successfully treated CRC as stand-alone therapy. This effect depended on intratumoral infiltration and activation of CD8+ T cells. Single application of Roseburia intestinalis or Anaerostipes caccae was even more effective than CC4. In a direct comparison, the CC4 mix supplementation outperformed anti-PD-1 therapy in mouse models of CRC and melanoma. Our findings provide a strong preclinical foundation for exploring gut bacteria as novel stand-alone therapy against solid tumors.


Asunto(s)
Terapia Biológica , Clostridiales/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Microbioma Gastrointestinal , Animales , Linfocitos T CD8-positivos/inmunología , Clostridiales/fisiología , Neoplasias Colorrectales/microbiología , Humanos , Inmunidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Simbiosis
11.
Adv Healthc Mater ; 10(16): e2100536, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34137204

RESUMEN

A limited infiltration and the subsequent low effective drug concentration result in poor chemotherapeutic outcomes against tumors, and even further promote tumor resistance and metastatic. Herein, iRGD-modified graphene oxide (GO) nanosheets (IPHG) are developed for the intensive treatment of metastatic tumors using focus-specific penetrated delivery together with photothermal therapy-mediated chemosensitization and photothermal therapy-strengthened integrin targeting-based antimigration. In vitro and in vivo data verified the mechanism of the tumor-selective infiltration of IPHG is based on a rigid 2D structure-associated advantage regarding hemodynamics and endothelial contact, followed by iRGD-endowed transendothelial and intratumoral transport. Once IPHG-DOX-penetrated 4T1 tumors are exposed to near-infrared irradiation, hyperthermia stress and photothermal therapy-elevated effective drug concentrations result in chemosensitization and prominent tumor suppression. Meanwhile, the specific binding of iRGD to integrins and photothermal therapy leads to the synergistic perturbation of cytoskeleton remodeling and subsequent impairment of cell motility and metastasis. The tailored design of IPHG validates a promising paradigm for drug delivery to combat tumor resistance and metastasis resulting from poor target access for single chemotherapy.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Línea Celular Tumoral , Doxorrubicina , Grafito , Humanos , Integrinas , Neoplasias/tratamiento farmacológico , Fototerapia
12.
Adv Sci (Weinh) ; 8(15): e2100233, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34085419

RESUMEN

Aberrant mechanical properties and immunosuppression are the two key factors that limit the antitumor efficacy of T cell immune checkpoint blockade inhibitors, e.g., programmed cell death-1 antibody (PD-1 Ab), against solid tumors in the clinic. This study leverages hyperbaric oxygen (HBO) for the first time to address these two issues and reports the PD-1-Ab-mediated immune responses against various stroma-rich solid malignancies. The results demonstrate that HBO promoted PD-1 Ab delivery and T cells infiltration into tumor parenchyma by depleting the extracellular matrix's main components, such as collagen and fibronectin. Furthermore, HBO disrupts hypoxia-mediated immunosuppression and helps PD-1 Ab trigger robust cytotoxic T lymphocytes and long-lasting immunological memory to inhibit tumor relapses. Such enhanced immune responses are effective in solid tumors from rodents and the cancer cells from hepatocellular carcinoma patients. The results illustrate that HBO bolsters antitumor efficacy of PD-1 Ab, and the HBO-PD-1 Ab combination is a promising stroma-rich solid tumors' treatment in the clinic.


Asunto(s)
Oxigenoterapia Hiperbárica/métodos , Inmunidad/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C
13.
Int J Mol Sci ; 22(11)2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-34072732

RESUMEN

CAR-T (chimeric antigen receptor T) cells have emerged as a milestone in the treatment of patients with refractory B-cell neoplasms. However, despite having unprecedented efficacy against hematological malignancies, the treatment is far from flawless. Its greatest drawbacks arise from a challenging and expensive production process, strict patient eligibility criteria and serious toxicity profile. One possible solution, supported by robust research, is the replacement of T lymphocytes with NK cells for CAR expression. NK cells seem to be an attractive vehicle for CAR expression as they can be derived from multiple sources and safely infused regardless of donor-patient matching, which greatly reduces the cost of the treatment. CAR-NK cells are known to be effective against hematological malignancies, and a growing number of preclinical findings indicate that they have activity against non-hematological neoplasms. Here, we present a thorough overview of the current state of knowledge regarding the use of CAR-NK cells in treating various solid tumors.


Asunto(s)
Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Técnicas de Cultivo de Célula , Ensayos Clínicos como Asunto , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Humanos , Células Asesinas Naturales/metabolismo , Neoplasias/diagnóstico , Neoplasias/etiología , Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Resultado del Tratamiento
14.
Zhonghua Yi Xue Za Zhi ; 100(42): 3323-3327, 2020 Nov 17.
Artículo en Chino | MEDLINE | ID: mdl-33202495

RESUMEN

Objective: To explore the clinical characteristics of acute myeloid leukemia (AML) complicated with simultaneous multiple primary cancer (SMPC). Methods: The data of 12 AML patients with SMPC hospitalized in the Affiliated Cancer Hospital of Zhengzhou University, the First Affiliated Hospital of Nanyang Medical College, the Xinhua District Hospital of Pingdingshan City and the First People's Hospital of Pingdingshan City from March 2014 to July 2019 were analyzed retrospectively, and their clinical features, treatment and prognosis were summarized. Results: Among the 12 patients, there were 6 males and 6 females, with a median age of 58 years (39-70 years). AML classification: according to French-American-British (FAB) classification, the 12 AML patients were classified as M0 1, M1 1, M2a 5, M2b 1, M3 2, M5 2; according to National Comprehensive Cancer Network (NCCN) prognosis stratified, low risk group 1 case, medium risk group 4 cases, high risk group 7 cases; classification of solid tumors: 3 cases of lung cancer, 1 case of breast cancer, 2 cases of gastric cancer, 3 cases of esophageal cancer, 1 case of rectal neuroendocrine tumor, 1 case of invasive hydatidiform mole and 1 case of sigmoid colon cancer. The median time interval for the diagnosis of two primary malignant tumors was 4 (from 2.6 to 5.6) months. Results of gene mutation detection: AML prognostic gene detection results: a total of 12 kinds of gene abnormalities including ASXL1, JAK2, TET2, U2AF1, ABCB1, FLT3-ITD, RUNX1, SETBPIT, TET2 (single nucleotide polymorphism, SNP), p53, IKZF1 and IDH2 were detected, and solid tumor related genes were detected: a total of 4 kinds of gene abnormalities including Her-2, EGFR, K-RAS and MSI were detected. Survival: among the 12 patients, 1 case was lost during follow-up, 2 cases were still in treatment, 3 cases ended treatment and the condition was stable, 6 cases died. The median overall survival of 12 patients was 12.5 (from 3.8 to 48.0) months. Conclusions: It is not clear whether there is a certain correlation between the simultaneous occurrence of AML and solid tumors. Patients with AML and synchronous solid tumors are not unusual. Both tumors should be treated aggressively at the same time.


Asunto(s)
Leucemia Mieloide Aguda , Neoplasias Primarias Múltiples , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Pronóstico , Estudios Retrospectivos
15.
Cancer Chemother Pharmacol ; 86(5): 673-679, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33030582

RESUMEN

PURPOSE: Hypomagnesemia has been associated with febrile neutropenia (FN) in pediatric patients receiving cisplatin-based chemotherapy (CDDPBC). The primary aim was to determine whether oral magnesium supplementation reduces FN episodes in pediatric patients with solid tumors treated with CDDPBC. METHOD: This randomized clinical trial, with open-label, single-center, parallel group and superiority design was conducted in Hospital Infantil de Mexico Federico Gomez at Mexico City. Children ≥ 9 years with solid tumors that were to receive a CDDPBC cycle were invited to participate. Each chemotherapy cycle with CDDPBC was randomly assigned to receive oral magnesium supplementation (250 mg/day) or not receive magnesium supplementation (control group). Efficacy was determined by relative risks (RR) with 95% confidence intervals (95% CI) as well as with numbers needed to treat (NNT). Active surveillance was conducted to assess safety in both groups. Analyses were carried out by intention to treat. ClinicalTrials.gov number NCT03449693. RESULTS: One hundred and one chemotherapy cycles with CDDPBC were analyzed (50 in the magnesium supplement arm and 51 in control group). Baseline clinical characteristics were similar comparing both groups. Oral magnesium supplementation reduces FN episodes compared to control group [RR 0.53, (95% CI 0.32-0.89), NNT = 4]. In the supplemented group, patients had fewer episodes of septic shock secondary to FN [RR 0.43, (95% CI 0.02-0.94), NNT = 6] and FN appeared on average 5 days later (p = 0.031). Hypomagnesemia episodes and adverse events were similar across both groups. CONCLUSION: Oral supplementation with magnesium reduces FN episodes neutropenia in pediatric patients with solid tumors treated with CDDPBC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Suplementos Dietéticos/efectos adversos , Neutropenia Febril/prevención & control , Magnesio/administración & dosificación , Neoplasias/tratamiento farmacológico , Administración Oral , Adolescente , Niño , Cisplatino/efectos adversos , Neutropenia Febril/epidemiología , Neutropenia Febril/etiología , Filgrastim/administración & dosificación , Estudios de Seguimiento , Humanos , Magnesio/efectos adversos , Masculino , México
16.
Cancer Med ; 9(14): 4991-5007, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32436621

RESUMEN

BACKGROUND: Treatment of patients with solid tumors and KRAS mutations remains disappointing. One option is the combined inhibition of pathways involved in RAF-MEK-ERK and PI3K-AKT-mTOR. METHODS: Patients with relapsed solid tumors were treated with escalating doses of everolimus (E) 2.5-10.0 mg/d in a 14-day run-in phase followed by combination therapy with sorafenib (S) 800 mg/d from day 15. KRAS mutational status was assessed retrospectively in the escalation phase. Extension phase included KRAS-mutated non-small-cell lung cancer (NSCLC) only. Pharmacokinetic analyses were accompanied by pharmacodynamics assessment of E by FDG-PET. Efficacy was assessed by CT scans every 6 weeks of combination. RESULTS: Of 31 evaluable patients, 15 had KRAS mutation, 4 patients were negative for KRAS mutation, and the KRAS status remained unknown in 12 patients. Dose-limiting toxicity (DLT) was not reached. The maximum tolerated dose (MTD) was defined as 7.5 mg/d E + 800 mg/d S due to toxicities at previous dose level (10 mg/d E + 800 mg/d S) including leucopenia/thrombopenia III° and pneumonia III° occurring after the DLT interval. The metabolic response rate in FDG-PET was 17% on day 5 and 20% on day 14. No patient reached partial response in CT scan. Median progression free survival (PFS) and overall survival (OS) were 3.25 and 5.85 months, respectively. CONCLUSIONS: Treatment of patients with relapsed solid tumors with 7.5 mg/d E and 800 mg/d S is safe and feasible. Early metabolic response in FDG-PET was not confirmed in CT scan several weeks later. The combination of S and E is obviously not sufficient to induce durable responses in patients with KRAS-mutant solid tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Everolimus/uso terapéutico , Fluorodesoxiglucosa F18/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Tomografía de Emisión de Positrones/métodos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Sorafenib/uso terapéutico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Everolimus/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sorafenib/farmacología
17.
Biofactors ; 46(4): 514-530, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32445262

RESUMEN

As an evolutionarily conserved pathway, Hippo signaling pathway impacts different pathology and physiology processes such as wound healing, tissue repair/size and regeneration. When some components of Hippo signaling dysregulated, it affects cancer cells proliferation. Moreover, the relation Hippo pathway with other signaling including Wnt, TGFß, Notch, and EGFR signaling leaves effect on the proliferation of cancer cells. Utilizing a number of therapeutic approaches, such as siRNAs and long noncoding RNA (lncRNA) to prevent cancer cells through the targeting of Hippo pathways, can provide new insights into cancer target therapy. The purpose of present review, first of all, is to demonstrate the importance of Hippo signaling and its relation with other signaling pathways in cancer. It also tries to demonstrate targeting Hippo signaling progress in cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/terapia , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/efectos de los fármacos , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , MicroARNs/metabolismo , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Notch/antagonistas & inhibidores , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
18.
AJR Am J Roentgenol ; 213(6): 1232-1239, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31613663

RESUMEN

OBJECTIVE. The purpose of this study was to investigate whether, compared with traditional criteria, the modified Response Evaluation Criteria in Solid Tumors version 1.1 for immune-based therapeutics (iRECIST) improves prediction of local tumor control and survival in patients with hepatocellular carcinoma (HCC) treated with stereotactic body radiotherapy (SBRT). MATERIALS AND METHODS. Fifty-one HCC lesions (mean size, 3.1 cm) treated with SBRT in 41 patients (mean age, 67 years) were retrospectively included. Each patient underwent CT or MRI before SBRT and at least once after SBRT. Best overall response was categorized using Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), iRECIST, World Health Organization (WHO) criteria, modified Response Evaluation Criteria in Solid Tumors (mRECIST), and European Association for the Study of the Liver (EASL) criteria. Lesions were then classified as local tumor control (i.e., stable disease, partial response, or complete response) or local treatment failure (i.e., progressive disease) by each tumor response criteria. Proportions of local tumor control were compared using the McNemar exact test. The 1-year overall survival was estimated using the Kaplan-Meier method. RESULTS. The median follow-up after SBRT was 21.0 months. The local tumor control rate was 94.1% (48/51) by iRECIST, 88.2% (45/51) by RECIST 1.1, 72.5% (37/51) by WHO criteria, 80.4% (41/51) by mRECIST, and 72.5% (37/51) by EASL criteria. The local tumor control rate was significantly higher according to iRECIST compared with WHO (p = 0.0010) and EASL (p = 0.0225) criteria. The 1-year survival rate for patients with local tumor control according to iRECIST (86.4%) was higher (although not statistically significant) compared with the 1-year survival rate for patients with local tumor control according to the other response criteria. CONCLUSION. iRECIST may provide more robust interpretation of HCC response after SBRT, yielding improved prediction of local tumor control and 1-year survival rates compared with traditional criteria.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/radioterapia , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/radioterapia , Criterios de Evaluación de Respuesta en Tumores Sólidos , Anciano , Anciano de 80 o más Años , Medios de Contraste , Femenino , Humanos , Masculino , Persona de Mediana Edad , Radiocirugia , Estudios Retrospectivos , Tasa de Supervivencia , Resultado del Tratamiento
19.
Bioorg Med Chem ; 27(1): 208-215, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30528162

RESUMEN

We previously demonstrated that capsazepine (CPZ), a synthetic transient receptor potential Vanilloid subtype 1 (TRPV1) antagonist, has significant anti-cancer effects in vivo. The purpose of this study was to develop more potent analogs based upon CPZ pharmacophore and structure-activity relationships (SAR) across analogs. We generated 30 novel compounds and screened for their anti-proliferative effects in cultured HeLa cervical cancer cells. Cell viability assays identified multiple compounds with IC50s < 15 µM and one compound, 29 with an IC50 < 5 µM; six fold more potent than CPZ. We validated the anti-proliferative efficacy of two lead compounds, 17 and 29, in vivo using HeLa-derived xenografts in athymic nude mice. Both analogs significantly reduced tumor volumes by day 8 compared to control treated animals (p < 0.001) with no observable adverse effects. Calcium imaging determined that compound 17 activates TRPV1 whereas 29 neither activates nor inhibits TRPV1; indicating a unique mechanism-of-action that does not involve TRPV1 signaling. Cell viability assays using a panel of additional tumor types including oral squamous cell carcinoma, non-small cell lung cancer (NSCLC), breast cancer, and prostate cancer cell lines (HSC-3, H460, MDA-231, and PC-3 respectively) demonstrated that both lead compounds were efficacious against every cancer type tested. Compounds 29 displayed IC50s of 1-2.5 µM in HSC-3and PC-3cells. Thus, we propose that these novel CPZ analogs may serve as efficacious therapeutic agents against multiple tumor types that warrant further development for clinical application.


Asunto(s)
Antineoplásicos/uso terapéutico , Capsaicina/análogos & derivados , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Capsaicina/síntesis química , Capsaicina/farmacología , Capsaicina/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones Desnudos , Estructura Molecular , Relación Estructura-Actividad , Canales Catiónicos TRPV/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Oncol Pharm Pract ; 25(1): 192-197, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29587607

RESUMEN

PURPOSE: Report bleeding incidences associated with rivaroxaban in adult patients with solid tumor malignancies requiring anticoagulation therapy. METHODS: This retrospective review was conducted at Indiana University Health, University Hospital and the Simon Cancer Center in Indianapolis, IN from January 2013 - February 2016. Patients were included if they had a solid tumor malignancy and prescribed rivaroxaban. Data were collected on 144 patients. Major bleeding was defined as bleeding requiring treatment (local, systemic treatment, blood cell transfusions) or hospitalization and minor bleeding was defined as bleeding not requiring treatment or hospitalization. RESULTS: Sixty-four (44%) patients experienced bleeding while on rivaroxaban. There were six cancer types that had a higher incidence of bleeding: bladder, breast, melanoma, pancreas, prostate, and renal cell cancers; 40% (6/15) of patients with bladder cancer experienced bleeding; 54% (7/13) with breast cancer experienced bleeding; 40% (4/10) of patients with melanoma experienced bleeding; 58% (11/19) of patients with pancreatic cancer experienced bleeding; 45% (10/22) of patients with prostate cancer experienced bleeding; and 56% (5/9) of patients with renal cell carcinoma experienced bleeding. No other data collected identified increased incidence of bleeding. CONCLUSIONS: Patients on rivaroxaban with a diagnosis of bladder, breast, melanoma, pancreas, prostate, or renal cell cancers had a higher incidence of bleeding compared to other solid tumors. Major bleeding was higher in bladder, breast, pancreas, and renal cell carcinomas, while minor bleeding was higher in patients with melanoma and prostate cancer.


Asunto(s)
Inhibidores del Factor Xa/efectos adversos , Hemorragia/inducido químicamente , Hemorragia/epidemiología , Neoplasias/tratamiento farmacológico , Neoplasias/epidemiología , Rivaroxabán/efectos adversos , Adulto , Anciano , Anticoagulantes/efectos adversos , Registros Electrónicos de Salud/tendencias , Femenino , Hospitalización/tendencias , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA