Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 342
Filtrar
1.
Environ Toxicol ; 39(7): 4058-4065, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38661261

RESUMEN

Platinum-based antineoplastic drugs, including cisplatin, carboplatin, and oxaliplatin, are widely used in the treatment of various cancers. Ototoxicity is a common adverse effect of platinum-based drugs. Ototoxicity leads to irreversible hearing impairment. We hypothesize that different platinum-based drugs exhibit varying ototoxic concentrations, time effects, and ototoxic mechanisms. We tested this hypothesis by using a zebrafish model (pvalb3b: TagGFP) to assess the viability of hair cells collected from zebrafish larvae. Cisplatin, carboplatin, and oxaliplatin were administered at dosages of 100, 200, or 400 µM, and the ototoxic effects of these drugs were assessed 1, 2, or 3 h after administration. Fm4-64 and a TUNEL assay were used to label the membranes of living hair cells and to detect cell apoptosis, respectively. We observed that >50% of hair cells were damaged at 1 h after cisplatin (100 µM) exposure, and this ototoxic effect increased at higher dosages and over time. Owing to the smaller ototoxic effects of carboplatin and oxaliplatin, we conducted higher-strength and longer-duration experiments with these drugs. Neither carboplatin nor oxaliplatin was obviously ototoxic, even at 1600 µM and after 6 h. Moreover, only cisplatin damaged the membranes of the hair cells. Cell apoptosis and significantly increased antioxidant gene expression were observed in only the cisplatin group. In conclusion, cisplatin significantly damages sensory hair cells and has notable dosage and time effects. Carboplatin and oxaliplatin are less ototoxic than cisplatin, likely due to having different ototoxic mechanisms than cisplatin.


Asunto(s)
Antineoplásicos , Apoptosis , Carboplatino , Cisplatino , Ototoxicidad , Oxaliplatino , Pez Cebra , Animales , Cisplatino/toxicidad , Oxaliplatino/toxicidad , Carboplatino/toxicidad , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Células Ciliadas Auditivas/efectos de los fármacos , Larva/efectos de los fármacos
2.
Toxicol Appl Pharmacol ; 481: 116751, 2023 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-37944569

RESUMEN

BACKGROUND: Cisplatin, carboplatin, and oxaliplatin are the only three platinum-based antineoplastic drugs that have been accepted worldwide for treating various cancers. Up to 83.6% of patients treated with platinum-based antineoplastic drugs will develop chemotherapy-induced peripheral neuropathy (CIPN), manifesting as sensory paresthesias, dysesthesias, and hypoesthesias that can cause significant adverse impact to daily activities. AIM: To investigate how these three platinum-based drugs affect mitochondrial function and myelination state of Schwann cells and the signalling pathway involved. METHOD: 2 µM Cisplatin, 20 µM carboplatin, and 1 µM oxaliplatin were used to inhibit the growth of CAL-27 by 20% respectively. These drugs were then used to induce chemotherapy-induced peripheral neuropathy in Rat Schwann Cells (RSC96). The changes in cell metabolism and myelin formation in RSC96 were investigated. RESULT: Cisplatin and carboplatin, but not oxaliplatin increased intracellular and mitochondrial reactive oxygen species in RSC96. Only Cisplatin and carboplatin decreased mitochondrial membrane potential (ΔΨm) and ATP production in RSC96. Both Cisplatin and carboplatin led to demyelination of RSC96, characterized by increased expression of p75NTR and decreased expression of myelin protein zero (MPZ). CONCLUSION: Cisplatin and carboplatin, but not oxaliplatin, caused mitochondrial dysfunction and induced demyelination in RSC96 while showing similar toxicity to head and neck cancer cells. Oxaliplatin may be a potential chemotherapy drug to prevent CIPN in patients with head and neck cancer.


Asunto(s)
Antineoplásicos , Carcinoma de Células Escamosas , Enfermedades Desmielinizantes , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Enfermedades del Sistema Nervioso Periférico , Humanos , Ratas , Animales , Cisplatino/farmacología , Carboplatino/toxicidad , Oxaliplatino/efectos adversos , Platino (Metal)/efectos adversos , Carcinoma de Células Escamosas de Cabeza y Cuello/inducido químicamente , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Antineoplásicos/toxicidad , Células de Schwann , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades Desmielinizantes/inducido químicamente
3.
Environ Health ; 22(1): 87, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38098045

RESUMEN

BACKGROUND: Exposure to per- and poly-fluoroalkyl substances (PFAS) has been associated with significant alterations in female reproductive health. These include changes in menstrual cyclicity, timing of menarche and menopause, and fertility outcomes, as well as increased risk of endometriosis, all of which may contribute to an increased risk of endometrial cancer. The effect of PFAS on endometrial cancer cells, specifically altered treatment response and biology, however, remains poorly studied. Like other gynecologic malignancies, a key contributor to lethality in endometrial cancer is resistance to chemotherapeutics, specifically to platinum-based agents that are used as the standard of care for patients with advanced-stage and/or recurrent disease. OBJECTIVES: To explore the effect of environmental exposures, specifically PFAS, on platinum-based chemotherapy response and mitochondrial function in endometrial cancer. METHODS: HEC-1 and Ishikawa endometrial cancer cells were exposed to sub-cytotoxic nanomolar and micromolar concentrations of PFAS/PFAS mixtures and were treated with platinum-based chemotherapy. Survival fraction was measured 48-h post-chemotherapy treatment. Mitochondrial membrane potential was evaluated in both cell lines following exposure to PFAS ± chemotherapy treatment. RESULTS: HEC-1 and Ishikawa cells displayed differing outcomes after PFAS exposure and chemotherapy treatment. Cells exposed to PFAS appeared to be less sensitive to carboplatin, with instances of increased survival fraction, indicative of platinum resistance, observed in HEC-1 cells. In Ishikawa cells treated with cisplatin, PFAS mixture exposure significantly decreased survival fraction. In both cell lines, increases in mitochondrial membrane potential were observed post-PFAS exposure ± chemotherapy treatment. DISCUSSION: Exposure of endometrial cancer cell lines to PFAS/PFAS mixtures had varying effects on response to platinum-based chemotherapies. Increased survival fraction post-PFAS + carboplatin treatment suggests platinum resistance, while decreased survival fraction post-PFAS mixture + cisplatin exposure suggests enhanced therapeutic efficacy. Regardless of chemotherapy sensitivity status, mitochondrial membrane potential findings suggest that PFAS exposure may affect endometrial cancer cell mitochondrial functioning and should be explored further.


Asunto(s)
Neoplasias Endometriales , Fluorocarburos , Femenino , Humanos , Carboplatino/toxicidad , Carboplatino/uso terapéutico , Cisplatino/farmacología , Cisplatino/uso terapéutico , Platino (Metal)/uso terapéutico , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/inducido químicamente , Línea Celular
4.
Drug Chem Toxicol ; 45(5): 2146-2152, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33829940

RESUMEN

Carboplatin is amongst the most commonly used anticancer drugs for the management of several human malignancies. However, it has displayed genotoxic properties against normal cells. Evaluation of natural products for their protective effects against chemotherapeutic drug induced toxicity has been growing in recent years. A naturally occurring flavonoid, chrysin, has strong antioxidant abilities and protects against DNA impairment. This study used multiple assays to evaluate the levels of damage to DNA in normal cells and to examine any possible protective role of chrysin against such damage. Male BALB/c mice were administered chrysin orally in two doses of 20 and 40 mg/kg for 10 consecutive days and then a single injection of carboplatin [90 mg/kg body weight (b.w.)] was administered intraperitoneally to induce carboplatin toxicity. 24 h after the carboplatin injection, mice were sacrificed. DNA damage was evaluated using several genotoxicity tests (8-Hydroxydeoxy-guanosine marker, comet assay, micronucleus test, and chromosomal aberration assay) to identify diverse types of damage to the DNA. The results suggest that pretreatment with chrysin significantly decreased the level of DNA damage caused by carboplatin probably due to its potent antioxidant traits. Therefore, chrysin can be considered to be developed as a chemoprotective agent against chemotherapy associated side-effects.


Asunto(s)
Antioxidantes , Daño del ADN , Animales , Antioxidantes/farmacología , Carboplatino/toxicidad , Ensayo Cometa/métodos , ADN , Flavonoides/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Micronúcleos
5.
Vet Surg ; 50(8): 1650-1661, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34375028

RESUMEN

OBJECTIVE: Evaluate local tissue toxicity and plasma platinum (Pt) in vivo after subcutaneous implantation of carboplatin-impregnated calcium sulfate hemihydrate (CI-CSH) beads. STUDY DESIGN: In vivo experimental study. ANIMALS: Eight male Sprague-Dawley rats. METHODS: CI-CSH beads were implanted subcutaneously (5 mg carboplatin/rat; 13.5 mg/kg carboplatin; 7.08 mg/kg Pt; 1.18 mg/m2 Pt) in eight rats (d0). Wound healing (daily), radiographic bead dissolution (weekly), systemic Pt uptake (plasma-Pt), local tissue Pt (d28), and histologic changes compared to nonincised and incised catheterization sites (d28) were assessed. Blood and tissue samples were analyzed by inductively coupled plasma mass spectrometry for Pt, and pharmacokinetic analysis was performed using noncompartmental methods. RESULTS: One rat died at d10, the remainder survived until d28. No wound complications were seen. The CI-CSH implantation site had higher histopathology scores than the other sites for necrosis (p = .013) and fibrosis (p = .013). Beads decreased in density radiographically (d0 to d28) (p = .062). Peak plasma-Pt concentration was 225.78 ng/ml at 12 h, and decreased over time, but Pt was still detectable on d28. The elimination half-life was 5.03 ± 1.13 days. Only 1.69% of implanted Pt remained in the beads at d28. CONCLUSIONS: CI-CSH beads incited microscopic mild inflammation but wound healing was not impaired. Pt was absorbed systemically and the release from the beads was near complete at d28. CLINICAL SIGNIFICANCE: Piled CI-CSH bead implantation is well tolerated in rats with similar elution profile as previously described. Beads were radiographically visible at d28. Minimal Pt was detected systemically suggesting Pt release does not match bead dissolution.


Asunto(s)
Sulfato de Calcio , Platino (Metal) , Animales , Carboplatino/toxicidad , Masculino , Ratas , Ratas Sprague-Dawley
6.
Toxicol Mech Methods ; 31(2): 138-149, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33190582

RESUMEN

Colon cancer is the commonest cancer worldwide. α-Hederin is a monodesmosidic triterpenoid saponin possessing diverse pharmacological activities. The running experiment was designed to test the chemopreventive activity of α-hederin when used as an adjuvant to carboplatin in an experimental model of mouse colon hyperplasia induced by 1,2-dimethylhydrazine (DMH). Fifty male Swiss albino mice were classified into five groups: group (I): saline group, group (II): DMH-induced colon hyperplasia control group, group (III): DMH + carboplatin (5 mg/kg) group, group (IV): DMH + α-hederin (80 mg/kg) group, and group (V): DMH + carboplatin (5 mg/kg)+α-hederin (80 mg/kg) group. Analyzing of colonic tissue indicated that the disease control group showed higher colon levels of phospho-PI3K to total-PI3K, phospho-AKT to total-AKT and cyclin D1 concurrent with lower phospho-JNK/total JNK ratio and caspase 3. However, treatment with α-hederin, in combination with carboplatin, favorably ameliorated phosphorylation of PI3K/AKT/JNK proteins, increased colon caspase 3 and downregulated cyclin D1. Microscopically, α-hederin, in combination with carboplatin, produced the most reduction in the histologic hyperplasia score, enhanced the goblet cell survival in periodic acid Schiff staining and reduced proliferation (Ki-67 immunostaining) in the current colon hyperplasia model. Collectively, the current study highlighted for the first time that using α-hederin as an adjuvant to carboplatin enhanced its chemopreventive activity, improved JNK signaling and increased apoptosis. Hence, further studies are warranted to test α-hederin as a promising candidate with chemotherapeutic agents in treating colon cancer.


Asunto(s)
Neoplasias del Colon , Ácido Oleanólico , 1,2-Dimetilhidrazina , Animales , Apoptosis , Carboplatino/toxicidad , Colon/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/prevención & control , Hiperplasia/inducido químicamente , Hiperplasia/patología , Hiperplasia/prevención & control , Masculino , Ratones , Fosfatidilinositol 3-Quinasas
7.
Mol Hum Reprod ; 26(3): 129-140, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-31953538

RESUMEN

The treatment of childhood cancer with chemotherapy drugs can result in infertility in adulthood. Newer generations of drugs are developed to replace parent drugs, with the potential benefits of less toxic side effects. For platinum alkylating-like drugs, in contrast to the parent compound cisplatin, the newer-generation drug carboplatin is reported to have reduced toxicity in some respects, despite being administered at 5-15 times higher than the cisplatin dose. Whether carboplatin is also less toxic than cisplatin to the reproductive system is unknown. Here we compare the gonadotoxic impact of cisplatin and carboplatin on female and male mouse prepubertal gonads. In vitro cultured CD1 mouse ovaries or testis fragments were exposed to either cisplatin or carboplatin for 24 h on Day 2 of culture and analysed by Day 6. A dose response for each drug was determined for the ovary (0.5, 1 & 5 µg/ml cisplatin and 1, 5 & 10 µg/ml carboplatin) and the testis (0.01, 0.05 & 0.1 µg/ml cisplatin and 0.1, 0.5 & 1 µg/ml carboplatin). For the ovary, unhealthy follicles were evident from 1 µg/ml cisplatin (73% unhealthy, P = 0.001) and 5 µg/ml carboplatin (84% unhealthy, P = 0.001), with a concomitant reduction in follicle number (P = 0.001). For the testis, the proliferating germ cell population was significantly reduced from 0.05 µg/ml cisplatin (73% reduction, P = 0.001) and 0.5 µg/ml carboplatin (75% reduction, P = 0.001), with no significant impact on the Sertoli cell population. Overall, results from this in vitro animal model study indicate that, at patient equivalent concentrations, carboplatin is no less gonadotoxic than cisplatin.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , Carboplatino/toxicidad , Cisplatino/toxicidad , Ovario/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Femenino , Células de la Granulosa/efectos de los fármacos , Masculino , Ratones , Técnicas de Cultivo de Órganos , Folículo Ovárico/efectos de los fármacos , Ovario/química , Ovario/ultraestructura , Células de Sertoli/efectos de los fármacos , Maduración Sexual , Testículo/química , Testículo/ultraestructura
8.
Chem Res Toxicol ; 31(12): 1293-1304, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30381944

RESUMEN

Platinum drugs, including carboplatin and oxaliplatin, are commonly used chemotherapy drugs that kill cancer cells by forming toxic drug-DNA adducts. These drugs have a proven, but modest, efficacy against several aggressive subtypes of breast cancer but also cause several side effects that can lead to the cessation of treatment. There is a clinical need to identify patients who will respond to platinum drugs in order to better inform clinical decision making. Diagnostic microdosing involves dosing patients or patient samples with subtherapeutic doses of radiolabeled platinum followed by measurement of platinum-DNA adducts in blood or tumor tissue and may be used to predict patient response. We exposed a panel of six breast cancer cell lines to 14C-labeled carboplatin or oxaliplatin at therapeutic and microdose (1% therapeutic dose) concentrations for a range of exposure lengths and isolated DNA from the cells. The DNA was converted to graphite, and measurement of radiocarbon due to platinum-DNA adduction was quantified via accelerator mass spectrometry (AMS). We observed a linear correlation in adduct levels between the microdose and therapeutic dose, and the level of platinum-DNA adducts corresponded to cell line drug sensitivity for both carboplatin and oxaliplatin. These results showed a clear separation in adduct levels between the sensitive and resistant groups of cell lines that could not be fully explained or predicted by changes in DNA repair rates or mutations in DNA repair genes. Further, we were able to quantitate oxaliplatin-DNA adducts in the blood and tumor tissue of a metastatic breast cancer patient. Together, these data support the use of diagnostic microdosing for predicting patient sensitivity to platinum. Future studies will be aimed at replicating this data in a clinical feasibility trial.


Asunto(s)
Complejos de Coordinación/toxicidad , Aductos de ADN/análisis , Daño del ADN/efectos de los fármacos , Platino (Metal)/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carboplatino/química , Carboplatino/toxicidad , Línea Celular Tumoral , Complejos de Coordinación/química , Reparación del ADN/efectos de los fármacos , Femenino , Humanos , Espectrometría de Masas , Oxaliplatino/química , Oxaliplatino/toxicidad
9.
BMC Cancer ; 17(1): 407, 2017 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-28595616

RESUMEN

BACKGROUND: Ridaforolimus is a mammalian target of rapamycin inhibitor that has activity in solid tumors. Paclitaxel and carboplatin have broad antineoplastic activity in many cancers. This phase I trial was conducted to determine the safety profile, maximal tolerated dose, and recommended phase II dose and schedule of oral ridaforolimus combined with paclitaxel and carboplatin in patients with solid tumor cancers. METHODS: Eligible patients with advanced solid tumor cancers received oral 10 to 30 mg ridaforolimus daily for 5 consecutive days per week combined with intravenous paclitaxel (175 mg/m2) and carboplatin (area under the curve [AUC] 5-6 mg/mL/min) in 3-week cycles. A standard 3 + 3 design was used to escalate doses, with predefined changes to an alternate dosing schedule and/or changes in carboplatin AUC doses based on dose-limiting toxicity (DLT). Secondary information was collected regarding response and time to progression. Patients were continued on treatment if therapy was tolerated and if stable disease or better was demonstrated. RESULTS: Thirty-one patients were consented, 28 patients were screened, and 24 patients met eligibility requirements and received treatment. Two patients were replaced for events unrelated to drug-related toxicity, resulting in 22 DLT-evaluable patients. Two grade 4 DLTs due to neutropenia were observed at dose level 1. The next cohort was changed to a predefined alternate dosing schedule (days 1-5 and 8-12). DLTs were neutropenia, sepsis, mucositis, and thrombocytopenia. The most common adverse events were neutropenia, anemia, thrombocytopenia, fatigue, alopecia, nausea, pain, and leukopenia. Twenty-four patients received a median of 4 cycles (range, 1-12). Evaluable patients for response (n = 18) demonstrated a median tumor measurement decrease of 25%. The best response in these 18 patients included 9 patients with partial response (50%), 6 with stable disease (33%), and 3 with progressive disease (17%). Thirteen of these patients received treatment for 4 or more cycles. CONCLUSIONS: Treatment with ridaforolimus combined with paclitaxel and carboplatin had no unanticipated toxicities and showed antineoplastic activity. The recommended phase II dose and schedule is ridaforolimus 30 mg (days 1-5 and 8-12) plus day 1 paclitaxel (175 mg/m2) and carboplatin (AUC 5 mg/mL/min) on a 21-day cycle. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01256268 (trial registration date: December 1, 2010).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/efectos adversos , Dosis Máxima Tolerada , Neoplasias/tratamiento farmacológico , Paclitaxel/efectos adversos , Proyectos de Investigación , Sirolimus/análogos & derivados , Administración Oral , Adulto , Anciano , Anemia/inducido químicamente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Carboplatino/uso terapéutico , Carboplatino/toxicidad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Fatiga/inducido químicamente , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neutropenia/inducido químicamente , Paclitaxel/uso terapéutico , Paclitaxel/toxicidad , Sirolimus/administración & dosificación , Sirolimus/efectos adversos , Sirolimus/uso terapéutico , Sirolimus/toxicidad , Trombocitopenia/inducido químicamente
10.
J Pathol ; 237(2): 190-202, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25988668

RESUMEN

Recent studies reveal that chemotherapy can enhance metastasis due to host responses, such as augmented expression of adhesion molecules in endothelial cells and increased populations of myeloid cells. However, it is still unclear how tumour cells contribute to this process. Here, we observed that paclitaxel and carboplatin accelerated lung metastasis in tumour-bearing mice, while doxorubicin and fluorouracil did not. Mechanistically, paclitaxel and carboplatin induced similar changes in cytokine and angiogenic factors. Increased levels of CXCR2, CXCR4, S1P/S1PR1, PlGF and PDGF-BB were identified in the serum or primary tumour tissues of tumour-bearing mice treated by paclitaxel. The serum levels of CXCL1 and PDGF-BB and the tissue level of CXCR4 were also elevated by carboplatin. On the other hand, doxorubicin and fluorouracil did not induce such changes. The chemotherapy-induced cytokine and angiogenic factor changes were also confirmed in gene expression datasets from human patients following chemotherapy treatment. These chemotherapy-enhanced cytokines and angiogenic factors further induced angiogenesis, destabilized vascular integrity, recruited BMDCs to metastatic organs and mediated the proliferation, migration and epithelial-to-mesenchymal transition of tumour cells. Interestingly, inhibitors of these factors counteracted chemotherapy-enhanced metastasis in both tumour-bearing mice and normal mice injected intravenously with B16F10-GFP cells. In particular, blockade of the SDF-1α-CXCR4 or S1P-S1PR1 axes not only compromised chemotherapy-induced metastasis but also prolonged the median survival time by 33.9% and 40.3%, respectively. The current study delineates the mechanism of chemotherapy-induced metastasis and provides novel therapeutic strategies to counterbalance pro-metastatic effects of chemo-drugs via combination treatment with anti-cytokine/anti-angiogenic therapy.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Antineoplásicos/toxicidad , Carboplatino/toxicidad , Citocinas/metabolismo , Neoplasias Pulmonares/secundario , Melanoma Experimental/secundario , Paclitaxel/toxicidad , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Inhibidores de la Angiogénesis/farmacología , Proteínas Angiogénicas/antagonistas & inhibidores , Proteínas Angiogénicas/sangre , Animales , Becaplermina , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/antagonistas & inhibidores , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevención & control , Lisofosfolípidos/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/prevención & control , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Factor de Crecimiento Placentario , Proteínas Gestacionales/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptores CXCR4/metabolismo , Receptores de Interleucina-8B/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Células del Estroma/metabolismo , Células del Estroma/patología , Factores de Tiempo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Anal Bioanal Chem ; 408(9): 2309-18, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26825342

RESUMEN

The study of the distribution of the cytostatic drugs cisplatin, carboplatin, and oxaliplatin along the kidney may help to understand their different nephrotoxic behavior. Laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) allows the acquisition of trace element images in biological tissues. However, results obtained are affected by several variations concerning the sample matrix and instrumental drifts. In this work, an internal standardization method based on printing an Ir-spiked ink onto the surface of the sample has been developed to evaluate the different distributions and accumulation levels of the aforementioned drugs along the kidney of a rat model. A conventional ink-jet printer was used to print fresh sagittal kidney tissue slices of 4 µm. A reproducible and homogenous deposition of the ink along the tissue was observed. The ink was partially absorbed on top of the tissue. Thus, this approach provides a pseudo-internal standardization, due to the fact that the ablation sample and internal standard take place subsequently and not simultaneously. A satisfactory normalization of LA-ICP-MS bioimages and therefore a reliable comparison of the kidney treated with different Pt-based drugs were achieved even for tissues analyzed on different days. Due to the complete ablation of the sample, the transport of the ablated internal standard and tissue to the inductively coupled plasma-mass spectrometry (ICP-MS) is practically taking place at the same time. Pt accumulation in the kidney was observed in accordance to the dosages administered for each drug. Although the accumulation rate of cisplatin and oxaliplatin is high in both cases, their Pt distributions differ. The strong nephrotoxicity observed for cisplatin and the absence of such side effect in the case of oxaliplatin could explain these distribution differences. The homogeneous distribution of oxaliplatin in the cortical and medullar areas could be related with its higher affinity for cellular transporters such as MATE2-k.


Asunto(s)
Carboplatino/toxicidad , Cisplatino/toxicidad , Tinta , Riñón/efectos de los fármacos , Compuestos Organoplatinos/toxicidad , Impresión , Animales , Carboplatino/metabolismo , Cisplatino/metabolismo , Riñón/metabolismo , Compuestos Organoplatinos/metabolismo , Oxaliplatino , Ratas , Estándares de Referencia
12.
Chem Res Toxicol ; 28(12): 2250-2, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26544157

RESUMEN

This rapid report focuses on the pharmacodynamic mechanism of the carboplatin/paclitaxel combination and correlates it with its cytotoxicity. Consistent with the synergistic to additive antitumor activity (the combination index ranging from 0.53 to 0.94), cells exposed to this combination had significantly increased carboplatin-DNA adduct formation when compared to that of carboplatin alone (450 ± 30 versus 320 ± 120 adducts per 10(8) nucleotides at 2 h, p = 0.004). Removal of paclitaxel increased the repair of carboplatin-DNA adducts: 39.4 versus 33.1 adducts per 10(8) nucleotides per hour in carboplatin alone (p = 0.021). This rapid report provides the first pharmacodynamics data to support the use of carboplatin/paclitaxel combination in the clinic.


Asunto(s)
Carboplatino/metabolismo , Aductos de ADN/metabolismo , Paclitaxel/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Carboplatino/uso terapéutico , Carboplatino/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Aductos de ADN/uso terapéutico , Aductos de ADN/toxicidad , Sinergismo Farmacológico , Humanos , Paclitaxel/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
13.
J Pharmacokinet Pharmacodyn ; 42(6): 709-20, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26341875

RESUMEN

A mechanistic model describing the effects of chemotherapy and radiation on platelet counts and endogenous thrombopoietin (eTPO) in mice was developed. Thrombocytopenia was induced in mice by injection of carboplatin followed by the whole body irradiation on days 0, 28, and 56, with platelet and eTPO samples collected over 84 days. The pharmacodynamic model consisted of a series of aging compartments representing proliferating megakaryocyte precursors, megakaryocytes, and platelets with possible eTPO clearance through internalization. The cytotoxic effects of treatment were described by the kinetics of the effect (K-PD) model, and stimulation of platelet production by eTPO was considered to be driven by receptor occupancy. The proposed PD model adequately described the platelet counts and eTPO concentrations in mice by accounting for nadirs and peaks of platelet count, and rebounds in eTPO time course profiles. The estimates of model parameters were in good agreement with their physiological values reported in literature for mice with platelet lifespan of 4.3 days and 185 cMpl receptors per platelet. The predicted duration of the treatment effect was 0.82 h (approximately 5 carboplatin half-lives in mice). The data was not informative about the eTPO stimulatory effect as the nominal precursor production rate was sufficient to account for platelet response to treatment. The model quantified the inverse relationship between eTPO levels and platelet counts and offered an explanation of the tolerance effect observed in the eTPO data. The simulated rebound in free receptors levels correlated with rebounds in eTPO levels. The model suggests that the duration of the toxic effects is determined by the turnover of the proliferating cells in the bone marrow. This indicates that the lifespan of the target cells (megakaryocyte precursors, megakaryocytes and platelets) is a key determinant in the duration of both drug exposure and toxicity due to treatment. The model can be extended to account for pharmacokinetics of exogenous drugs and be applied to analysis of human data.


Asunto(s)
Antineoplásicos/toxicidad , Plaquetas/efectos de los fármacos , Plaquetas/efectos de la radiación , Carboplatino/toxicidad , Quimioradioterapia/efectos adversos , Modelos Biológicos , Modelos Estadísticos , Traumatismos por Radiación/inducido químicamente , Trombocitopenia/inducido químicamente , Irradiación Corporal Total/efectos adversos , Animales , Antineoplásicos/administración & dosificación , Biomarcadores/sangre , Plaquetas/metabolismo , Plaquetas/patología , Carboplatino/administración & dosificación , Simulación por Computador , Esquema de Medicación , Femenino , Humanos , Ratones , Recuento de Plaquetas , Dosis de Radiación , Traumatismos por Radiación/sangre , Receptores de Trombopoyetina/sangre , Medición de Riesgo , Trombocitopenia/sangre , Trombopoyesis/efectos de los fármacos , Trombopoyesis/efectos de la radiación , Trombopoyetina/sangre , Factores de Tiempo
14.
J Neurooncol ; 120(3): 507-13, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25108776

RESUMEN

The number of studies examining the use of tandem high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT) to treat high-risk or recurrent brain tumors is increasing. However, studies addressing the toxicity associated with tandem HDCT/auto-SCT, particularly during the second HDCT/auto-SCT, are very limited. For this reason, we retrospectively evaluated the toxicity of tandem HDCT/auto-SCT with carboplatin-thiotepa-etoposide (CTE) and cyclophosphamide-melphalan (CM) regimens when used to treat high-risk or recurrent brain tumors. A total of 109 patients who received a first HDCT/auto-SCT and 100 who proceeded to a second HDCT/auto-SCT between May 2005 and December 2013 were included. Hematologic recovery was rapid during both the first and second HDCT/auto-SCT. In the first HDCT/auto-SCT, mucositis-related gastrointestinal toxicity was frequent, and two (1.8 %) patients died from toxicity [one hepatic veno-occlusive disease (VOD) and one sepsis]. In the second HDCT/auto-SCT, mucositis-related toxicity was milder than in the first round. However, hepatic VOD frequency was high (20.0 %), and six (6.0 %) patients died from toxicity (four hepatic VODs, one asphyxia, and one sepsis). Multivariate analysis indicated that age younger than 8 years was the only significant predictor for hepatic VOD. All six patients who died from toxicity during the second HDCT/auto-SCT were younger than 9 years of age. This study demonstrates that tandem HDCT/auto-SCT using CTE/CM regimens was generally feasible. However, dose reduction during the second HDCT/auto-SCT in young children might be needed to decrease the death rate from toxicity.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , Neoplasias Encefálicas/terapia , Trasplante de Células Madre/efectos adversos , Adolescente , Adulto , Factores de Edad , Antineoplásicos Alquilantes/administración & dosificación , Carboplatino/administración & dosificación , Carboplatino/toxicidad , Niño , Preescolar , Terapia Combinada/efectos adversos , Etopósido/administración & dosificación , Etopósido/toxicidad , Femenino , Humanos , Lactante , Masculino , Melfalán/administración & dosificación , Melfalán/toxicidad , Estudios Retrospectivos , Tiotepa/administración & dosificación , Tiotepa/toxicidad , Trasplante Autólogo/efectos adversos , Adulto Joven
15.
Inorg Chem ; 53(7): 3371-84, 2014 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-24665859

RESUMEN

Bispidine (3,7-diazabicyclo[3.3.1]nonane, C7H14N2) analogues of cisplatin, carboplatin, and oxaliplatin have been prepared. (C7H14N2)PtCl2·DMF (1b), obtained from (1,5-hexadiene)PtCl2 and bispidine in DMF, is dimeric in the solid state. Dissolving 1b in hot N-methylformamide allows crystallization of the solvent-free polymeric (C7H14N2)PtCl2 (1a). Recrystallization of 1a,b from hot water yields the trihydrate (C7H14N2)PtCl2·3H2O (1c). Reaction of 1 with Ag2(cbdca) (cbdca = 1,1-cyclobutanedicarboxylate) in water affords the pentahydrate (C7H14N2)Pt{C4H6(CO2)2}·5H2O (2b), which loses water in vacuo to give (C7H14N2)Pt{C4H6(CO2)2} (2a). Reaction of 1 with AgNO3 in water, followed by addition of Na2C2O4, affords the water-free polymeric (C7H14N2)Pt(C2O4) (3). All complexes have been structurally characterized, revealing various patterns of N-H···Cl and N-H···O hydrogen bonds. In the hydrates 1c and 2b the complexes are embedded in intricate three-dimensional water networks. Complexes 1a, 2a, and 3 have been tested for their cytotoxicity against human cancer cell lines K562 (chronic myeloid leukemia), A2780 (ovarian cancer), and its platinum-resistant subline A2780 CisR and are compared to their parent analogues. The new complexes show significant cytotoxic activity along with a low platinum resistance factor.


Asunto(s)
Antineoplásicos/toxicidad , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Carboplatino/análogos & derivados , Carboplatino/toxicidad , Cisplatino/análogos & derivados , Cisplatino/toxicidad , Compuestos Organoplatinos/toxicidad , Antineoplásicos/síntesis química , Carboplatino/síntesis química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/síntesis química , Colorantes , Humanos , Modelos Moleculares , Conformación Molecular , Compuestos Organoplatinos/síntesis química , Oxaliplatino , Relación Estructura-Actividad , Sales de Tetrazolio , Tiazoles
16.
J Coll Physicians Surg Pak ; 33(7): 722-726, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37401209

RESUMEN

OBJECTIVE: To evaluate the histomorphological response of alpha-tocopherol co-administration with carboplatin chemotherapy. STUDY DESIGN: A laboratory-based experimental study. Place and Duration of the Study: Anatomy Department, Army Medical College / National University of Medical Sciences (NUMS), Rawalpindi, Pakistan, from January to December 2021. METHODOLOGY: Thirty adult Sprague-Dawley rats were divided into three groups of ten rats each. Control group A received normal diet and water, experimental group B was administered single injection of carboplatin 2.5 mg/Kg intraperitoneally; and experimental group C along with carboplatin injection also received alpha-tocopherol 62.7 mg/Kg daily. At the end of 12 weeks, the euthanasia of animals was done and kidneys were dissected out. Right-sided kidneys were stained with Haematoxylin and Eosin. Micrometry was done to measure the diameters of renal cortical tubules and renal corpuscles. RESULTS: The proximal and distal tubular and luminal diameter and transvertical diameter of renal corpuscle were increased in group B as compared to control group A. In group C, the proximal and distal tubular diameters were 5.175 ± 0.39 µm and 3.88 ± 0.364 µm, respectively; proximal and distal luminal diameters were 2.67 ± 0.35 µm and 1.64 ± 0.24 µm, respectively and transvertical diameter of renal corpuscle was 12.16 ± 0.870 µm. These values were less than experimental group B and closer to that of control group A. CONCLUSION: Renal microscopic parameters showed improvement in the group administered with alpha-tocopherol. Therefore, alpha-tocopherol has ameliorative effects on carboplatin-induced renal damage. KEY WORDS: Alpha-tocopherol, Carboplatin, Renal corpuscle, Tubules.


Asunto(s)
Riñón , alfa-Tocoferol , Ratas , Animales , alfa-Tocoferol/farmacología , Carboplatino/toxicidad , Ratas Sprague-Dawley , Antioxidantes/farmacología
17.
J Biol Inorg Chem ; 17(6): 891-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22638735

RESUMEN

Carboplatin, an analogue of "classical" cis-diamminedichloridoplatinum(II) (cisplatin), is a widely used second-generation platinum anticancer drug. Cytotoxicity of cisplatin and carboplatin is mediated by platinum-DNA adducts. Markedly higher concentrations of carboplatin are required, and the rate of adduct formation is considerably slower. The reduced toxic effects in tumor cells and a more acceptable side-effect profile are attributable to the lower reactivity of carboplatin with nucleophiles, since the cyclobutanedicarboxylate ligand is a poorer leaving group than the chlorides in cisplatin. Recently, platinum complexes were shown to be particularly attractive as potential photochemotherapeutic anticancer agents. Selective photoactivation of platinum complexes by irradiation of cancer cells may avoid enhancement of toxic side-effects, but may increase toxicity selectively in cancer cells and extend the application of photoactivatable platinum complexes to resistant cells and to a wider range of cancer types. Therefore, it was of interest to examine whether carboplatin can be affected by irradiation with light to the extent that its DNA binding and cytotoxic properties are altered. We have found that carboplatin is converted to species capable of enhanced DNA binding by UVA irradiation and consequently its toxicity in cancer cells is markedly enhanced. Recent advances in laser and fiber-optic technologies make it possible to irradiate also internal organs with light of highly defined intensity and wavelength. Thus, carboplatin is a candidate for use in photoactivated cancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/efectos de la radiación , Carboplatino/farmacología , Carboplatino/efectos de la radiación , ADN/química , Procesos Fotoquímicos/efectos de la radiación , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Sitios de Unión/efectos de los fármacos , Carboplatino/química , Carboplatino/toxicidad , Bovinos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Cinética , Plásmidos , Relación Estructura-Actividad , Células Tumorales Cultivadas , Rayos Ultravioleta
18.
Int J Clin Oncol ; 17(4): 367-72, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21850385

RESUMEN

BACKGROUND: Some regimens of chemotherapy cause peripheral neuropathy such as pain in muscles and joints and numbness in the limbs. It is often difficult to estimate the neuropathy accurately and analyze it in detail. The aim of this study was to investigate whether chemotherapy-induced peripheral neuropathy could be appropriately estimated by using the visual analogue scale (VAS). METHODS: Ninety-three patients who received paclitaxel and carboplatin treatment (TC) or paclitaxel and docetaxel treatment (DC) participated in answering a questionnaire about peripheral neuropathy using the VAS. As a result, 134 cycles of TC and 79 cycles of DC were evaluated. The average of VAS scores at every 10 days after each cycle of chemotherapy began was calculated. The daily change in VAS scores was also analyzed, and average VAS scores compared between TC and DC. RESULTS: Daily changes in peripheral neuropathy for each treatment could be demonstrated in detail. Pain and numbness had separate patterns of appearance. For both pain and numbness, a greater VAS score was observed in patients receiving TC than in those receiving DC. As the number of cycles grew, peripheral neuropathy became more serious in TC. CONCLUSIONS: The VAS could appropriately recognize the difference in peripheral neuropathy between TC and DC. Moreover, the VAS could also catch the change in peripheral neuropathy. This result suggests that the VAS system is a useful tool for managing peripheral neuropathy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Carboplatino , Paclitaxel , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Taxoides , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Carboplatino/administración & dosificación , Carboplatino/toxicidad , Docetaxel , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Paclitaxel/administración & dosificación , Paclitaxel/toxicidad , Dimensión del Dolor , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Encuestas y Cuestionarios , Taxoides/administración & dosificación , Taxoides/toxicidad
19.
J Assoc Res Otolaryngol ; 23(3): 379-389, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35378622

RESUMEN

Integration of acoustic information over time is essential for processing complex stimuli, such as speech, due to its continuous variability along the time domain. In both humans and animals, perception of acoustic stimuli is a function of both stimulus intensity and duration. For brief acoustic stimuli, as duration increases, thresholds decrease by approximately 3 dB for every doubling in duration until stimulus duration reaches 500 ms, a phenomenon known as temporal integration. Although hearing loss and damage to outer hair cells (OHC) have been shown to alter temporal integration in some studies, the role of cochlear inner hair cells (IHC) on temporal integration is unknown. Because IHC transmit nearly all acoustic information to the central auditory system and are believed to code both intensity and timing information, these sensory cells likely play a critical role in temporal integration. To test the hypothesis that selective IHC loss degrades the temporal integration function, behaviorally trained chinchillas were treated with carboplatin, a drug known to selectively destroy IHC with little to no effect on OHC in this species. Pure-tone thresholds were assessed across frequencies (1, 2, 4, 8, 12 kHz) as a function of signal duration (500, 100, 50, 10, and 5 ms). Baseline testing showed a significant effect of duration on thresholds. Threshold decreased as a function of increasing duration, as expected. Carboplatin treatment (75 mg/kg) produced a moderate to severe loss of IHC (45-85%) with little-to-no loss of OHC. Contrary to our hypothesis, post-carboplatin temporal integration thresholds showed no significant differences from baseline regardless of stimulus duration or frequency. These data suggest that few IHC are necessary for temporal integration of simple stimuli. Temporal integration may be sensitive to loss of OHC and loss of cochlear non-linearities but does not appear to be sensitive to selective IHC loss.


Asunto(s)
Células Ciliadas Auditivas Internas , Células Ciliadas Auditivas Externas , Animales , Umbral Auditivo , Carboplatino/toxicidad , Chinchilla , Cóclea
20.
Int J Nanomedicine ; 17: 3013-3041, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35836838

RESUMEN

Purpose: Ovarian cancer is the most lethal gynecologic malignancy. The combination of paclitaxel (PTX) and carboplatin (CBP) is the first-line remedy for clinical ovarian cancer. However, due to the limitations of adverse reaction and lacking of targeting ability, the chemotherapy of ovarian cancer is still poorly effective. Here, a novel estrone (ES)-conjugated PEGylated liposome co-loaded PTX and CBP (ES-PEG-Lip-PTX/CBP) was designed for overcoming the above disadvantages. Methods: ES-PEG-Lip-PTX/CBP was prepared by film hydration method and could recognize estrogen receptor (ER) over-expressing on the surface of SKOV-3 cells. The characterizations, stability and in vitro release of ES-PEG-Lip-PTX/CBP were studied. In vitro cellular uptake and its mechanism were observed by fluorescence microscope. In vivo targeting effect in tumor-bearing mice was determined. Pharmacokinetics and biodistribution were studied in ICR mice. In vitro cytotoxicity and in vivo anti-tumor efficacy were evaluated on SKOV-3 cells and tumor-bearing mice, respectively. Finally, the acute toxicity in ICR mice was explored for assessing the preliminary safety of ES-PEG-Lip-PTX/CBP. Results: Our results showed that ES-PEG-Lip-PTX/CBP was spherical shape without aggregation. ES-PEG-Lip-PTX/CBP exhibited the optimum targeting effect on uptake in vitro and in vivo. The pharmacokinetics demonstrated ES-PEG-Lip-PTX/CBP had improved the pharmacokinetic behavior. In vitro cytotoxicity showed that ES-PEG-Lip-PTX/CBP maximally inhibited SKOV-3 cell proliferation and its IC50 values was 1.6 times lower than that of non-ES conjugated liposomes at 72 h. The in vivo anti-tumor efficacy study demonstrated that ES-PEG-Lip-PTX/CBP could lead strong SKOV-3 tumor growth suppression with a tumor volume inhibitory rate of 81.8%. Meanwhile, acute toxicity studies confirmed that ES-PEG-Lip-PTX/CBP significantly reduced the toxicity of the chemo drugs. Conclusion: ES-PEG-Lip-PTX/CBP was successfully prepared with an optimal physicochemical and ER targeting property. The data of pharmacokinetics, anti-tumor efficacy and safety study indicated that ES-PEG-Lip-PTX/CBP could become a promising therapeutic formulation for human ovarian cancer in the future clinic.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Animales , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Carboplatino/uso terapéutico , Carboplatino/toxicidad , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Estrona/uso terapéutico , Femenino , Humanos , Liposomas/uso terapéutico , Ratones , Ratones Endogámicos ICR , Neoplasias Ováricas/tratamiento farmacológico , Paclitaxel/uso terapéutico , Polietilenglicoles/química , Distribución Tisular
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda