Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pediatr Blood Cancer ; 70(6): e30318, 2023 06.
Article in English | MEDLINE | ID: mdl-36973999

ABSTRACT

BACKGROUND: Successful engraftment of human cancer biopsies in immunodeficient mice correlates with the poor prognosis of patients. This was reported 30 years ago for children with neuroblastoma, but the standard of care treatment evolved significantly during the last 15 years, leading to improved survival of these patients. Here, we evaluated the association of patient-derived xenograft (PDX) engraftment and prognosis in patients receiving up-to-date treatments for cancers classified as metastatic (stage M) high-risk neuroblastoma (HR-NB) by the International Neuroblastoma Risk Group Staging System (INRGSS). METHODS: We obtained biopsies from patients with stage M HR-NB. We inoculated biopsy fragments subcutaneously in mice. We studied the association of PDX engraftment with event-free survival (EFS) and overall survival (OS) of patients. RESULTS: Since 2009, we established 17 PDX from 97 samples of 66 patients with stage M HR-NB, with a follow-up of at least two years. Factors associated with higher probability of engraftment were the death as outcome (p = .0006) and the amplification of the gene MYCN in tumors (p = .0271). Patients whose biopsies established a PDX had significantly shorter EFS and OS (p = .0039 and .0002, respectively) than patients whose samples did not engraft. The association of PDX engraftment and OS was significant in patients without MYCN amplification (p = .0041), but not in patients with MYCN amplification (p = .2707). CONCLUSION: Positive PDX engraftment is a factor related to poor prognosis and fatal outcome in patients with stage M HR-NB treated with up-to-date therapies.


Subject(s)
Neuroblastoma , Child , Humans , Animals , Mice , Infant , Prognosis , Heterografts , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/pathology , Progression-Free Survival , Gene Amplification , Neoplasm Staging
2.
Biochem Pharmacol ; 208: 115408, 2023 02.
Article in English | MEDLINE | ID: mdl-36603685

ABSTRACT

Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) shows potent preclinical anticancer activity in pediatric solid tumors such as Ewing sarcoma, rhabdomyosarcoma and neuroblastoma, but responses in clinical trials have been modest. In this work, we aimed to discover a rational biomarker-based approach to select the right candidate patients for this treatment. We assessed the efficacy of nab-paclitaxel in 27 patient-derived xenografts (PDX), including 14 Ewing sarcomas, five rhabdomyosarcomas and several other pediatric solid tumors. Response rate (partial or complete response) was remarkable in rhabdomyosarcomas (four of five) and Ewing sarcomas (four of 14). We addressed several predictive factors of response to nab-paclitaxel such as the expression of the secreted protein acidic and rich in cysteine (SPARC), chromosomal stability of cancer cells and expression of antiapoptotic members of the B-cell lymphoma-2 (Bcl-2) family of proteins such as Bcl-2, Bcl-xL, Bcl-W and Mcl-1. Protein (immunoblotting) and gene expression of SPARC correlated positively, while immunoblotting and immunohistochemistry expression of Bcl-2 correlated negatively with the efficacy of nab-paclitaxel in Ewing sarcoma PDX. The negative correlation of Bcl-2 immunoblotting signal and activity was especially robust (r = 0.8352; P = 0.0007; Pearson correlation). Consequently, we evaluated pharmacological strategies to inhibit Bcl-2 during nab-paclitaxel treatment. We observed that the Bcl-2 inhibitor venetoclax improved the activity of nab-paclitaxel in highly resistant Bcl-2-expressing Ewing sarcoma PDX. Overall, our results suggest that low Bcl-2 expression could be used to select patients with Ewing sarcoma sensitive to nab-paclitaxel, and Bcl-2 inhibitors could improve the activity of this drug in Bcl-2-expressing Ewing sarcoma.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Rhabdomyosarcoma , Sarcoma, Ewing , Child , Humans , Antineoplastic Agents/therapeutic use , Biomarkers , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Osteonectin/genetics , Osteonectin/metabolism , Osteonectin/therapeutic use , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Rhabdomyosarcoma/drug therapy , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/pathology
3.
J Pathol Clin Res ; 9(1): 32-43, 2023 01.
Article in English | MEDLINE | ID: mdl-36148636

ABSTRACT

Fundoscopy is the standard method for diagnosis and follow-up of intraocular retinoblastoma, but it is sometimes insufficient to discern whether tumors are inactivated following treatments. In this work, we hypothesized that the amount of conserved nuclear DNA sequences in the cell-free DNA (cfDNA) fraction of the aqueous humor (AH) might complement fundoscopy for retinoblastoma follow-up. To address our hypothesis, we developed highly sensitive droplet digital polymerase chain reaction (ddPCR) methods to quantify highly conserved DNA sequences of nucleus-encoded genes (GAPDH and B4GALNT1) and of a mitochondrial gene, MT-ATP6. We obtained AH samples during intravitreal treatments. We analyzed 42 AH samples from 25 patients with intraocular retinoblastoma and 11 AH from controls (non-cancer patients). According to clinical criteria, we grouped patients as having progression-free or progressive retinoblastoma. cfDNA concentration in the AH was similar in both retinoblastoma groups. Copy counts for nucleus-derived sequences of GAPDH and B4GALNT1 were significantly higher in the AH from patients with progressive disease, compared to the AH from progression-free patients and control non-cancer patients. The presence of mitochondrial DNA in the AH explained that both retinoblastoma groups had similar cfDNA concentration in AH. The optimal cut-off point for discriminating between progressive and progression-free retinoblastomas was 108 GAPDH copies per reaction. Among patients having serial AH samples analyzed during their intravitreal chemotherapy, GAPDH copies were high and decreased below the cut-off point in those patients responding to chemotherapy. In contrast, one non-responder patient remained with values above the cut-off during follow-up, until enucleation. We conclude that the measurement of conserved nuclear gene sequences in AH allows follow-up of intraocular retinoblastoma during intravitreal treatment. The method is applicable to all patients and could be relevant for those in which fundoscopy evaluation is inconclusive.


Subject(s)
Base Sequence , Humans
4.
J Pathol ; 257(3): 327-339, 2022 07.
Article in English | MEDLINE | ID: mdl-35254670

ABSTRACT

The microenvironment of retinoblastoma, the solid malignancy of the developing retina, is immunosuppressive. To study the interactions between tumor-associated microglia/macrophages (TAMs) and tumor cells in retinoblastomas, we analyzed immunohistochemistry markers in 23 patient samples and characterized 105 secreted cytokines of 11 retinoblastoma cell models in culture. We detected profuse infiltration of CD163+ protumoral M2-like polarized TAMs in eyes enucleated due to cancer progression. Previous treatment of patients increased the number of TAMs but did not affect M2-like polarization. M2-like microglia/macrophages were almost absent in five eyes obtained from children enucleated due to nontumoral causes. CD8+ tumor-infiltrating lymphocytes (TILs) were moderately abundant in tumor eyes and very scarce in nontumoral ones. The expression of the immune checkpoint molecule PD-L1 was absent in 95% of the tumor samples, which is concordant with the finding of FOXP3+ Tregs infiltrating tumors. We confirmed the pathology results using single-cell transcriptome analysis of one tumor. We identified the cytokines extracellular matrix metalloproteinase inducer (EMMPRIN) and macrophage migration inhibitory factor (MIF), both with reported immunosuppressive activity, secreted at high levels in retinoblastoma primary cell cultures. Gene expression analysis of a large retinoblastoma cohort and single-cell transcriptome analysis confirmed that MIF and EMMPRIN were significantly upregulated in retinoblastomas, which led us to quantify both proteins by immunoassays in liquid biopsies (aqueous humor obtained from more than 20 retinoblastoma patients). We found a significant increase in the concentration of MIF and EMMPRIN in cancer patients, compared to 12 noncancer ones. Finally, we showed that macrophages derived from peripheral blood mononuclear cells increased the expression of markers of M2-like polarization upon exposure to retinoblastoma-conditioned medium or recombinant MIF. Overall, our findings suggest that retinoblastoma cell secretions induce the protumoral phenotype of this tumor. Our results might have clinical impact in the fields of biomarkers and treatment. © 2022 The Pathological Society of Great Britain and Ireland.


Subject(s)
Retinal Neoplasms , Retinoblastoma , Aqueous Humor , Basigin , Humans , Leukocytes, Mononuclear , Retinal Neoplasms/genetics , Secretome , Tumor Microenvironment
5.
J Control Release ; 342: 81-92, 2022 02.
Article in English | MEDLINE | ID: mdl-34974029

ABSTRACT

Secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycoprotein overexpressed by several cancers. Because SPARC shows high binding affinity to albumin, we reasoned that pediatric sarcoma xenografts expressing SPARC would show enhanced uptake and accumulation of nanoparticle albumin-bound (nab)-paclitaxel, a potent anticancer drug formulation. We first evaluated the expression of SPARC in patient-derived xenografts (PDXs) of Ewing sarcoma, rhabdomyosarcoma and osteosarcoma, finding variable SPARC gene expression that correlated well with SPARC protein measured by immunoblotting. We revealed that the activity of the fusion gene chimera EWSR1-FLI1, the genetic driver of Ewing sarcoma, leads to lower expression of the gene SPARC in these tumors, likely due to enriched acetylation marks of the histone H3 lysine 27 at regions including the SPARC promoter and potential enhancers. Then, we used SPARC-edited Ewing sarcoma cells (A673 line) to demonstrate that SPARC knocked down (KD) cells accumulated significantly less amount of nab-paclitaxel in vitro than SPARC wild type (WT) cells. In vivo, SPARC KD and SPARC WT subcutaneous xenografts in mice achieved similar maximum intratumoral concentrations of nab-paclitaxel, though drug clearance from SPARC WT tumors was significantly slower. We confirmed such SPARC-mediated long-term intratumoral accumulation of nab-paclitaxel in Ewing sarcoma PDX with high expression of SPARC, which accumulated significantly more nab-paclitaxel than SPARC-low PDX. SPARC-high PDX responded better to nab-paclitaxel than SPARC-low tumors, although these results should be taken cautiously, given that the PDXs were established from different patients that could have specific determinants predisposing response to paclitaxel. In addition, SPARC KD Ewing sarcoma xenografts responded better to soluble docetaxel and paclitaxel than to nab-paclitaxel, while SPARC WT ones showed similar response to soluble and albumin-carried drugs. Overall, our results show that pediatric sarcomas expressing SPARC accumulate nab-paclitaxel for longer periods of time, which could have clinical implications for chemotherapy efficacy.


Subject(s)
Bone Neoplasms , Osteosarcoma , Albumins/metabolism , Animals , Bone Neoplasms/drug therapy , Humans , Mice , Osteonectin/genetics , Osteonectin/metabolism , Osteonectin/therapeutic use , Osteosarcoma/drug therapy , Paclitaxel/therapeutic use
6.
J Pathol Clin Res ; 7(4): 338-349, 2021 07.
Article in English | MEDLINE | ID: mdl-33837665

ABSTRACT

The goals of this work were to identify factors favoring patient-derived xenograft (PDX) engraftment and study the association between PDX engraftment and prognosis in pediatric patients with Ewing sarcoma, osteosarcoma, and rhabdomyosarcoma. We used immunodeficient mice to establish 30 subcutaneous PDX from patient tumor biopsies, with a successful engraftment rate of 44%. Age greater than 12 years and relapsed disease were patient factors associated with higher engraftment rate. Tumor type and biopsy location did not associate with engraftment. PDX models retained histology markers and most chromosomal aberrations of patient samples during successive passages in mice. Model treatment with irinotecan resulted in significant activity in 20 of the PDXs and replicated the response of rhabdomyosarcoma patients. Successive generations of PDXs responded similarly to irinotecan, demonstrating functional stability of these models. Importantly, out of 68 tumor samples from 51 patients with a median follow-up of 21.2 months, PDX engraftment from newly diagnosed patients was a prognostic factor significantly associated with poor outcome (p = 0.040). This association was not significant for relapsed patients. In the subgroup of patients with newly diagnosed Ewing sarcoma classified as standard risk, we found higher risk of relapse or refractory disease associated with those samples that produced stable PDX models (p = 0.0357). Overall, our study shows that PDX engraftment predicts worse outcome in newly diagnosed pediatric sarcoma patients.


Subject(s)
Prognosis , Sarcoma, Ewing/drug therapy , Xenograft Model Antitumor Assays/methods , Adolescent , Animals , Child , Child, Preschool , Disease Models, Animal , Female , Heterografts/drug effects , Humans , Irinotecan/pharmacology , Irinotecan/therapeutic use , Male , Mice , Osteosarcoma/drug therapy , Rhabdomyosarcoma/drug therapy , Sarcoma/drug therapy , Treatment Outcome
7.
Cancers (Basel) ; 13(4)2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33669730

ABSTRACT

Ewing's Sarcoma (ES) is the second most frequent malignant bone tumour in children and young adults and currently only untargeted chemotherapeutic approaches and surgery are available as treatment, although clinical trials are on-going for recently developed ES-targeted therapies. To study ES pathobiology and develop novel drugs, established cell lines and patient-derived xenografts (PDX) are the most employed experimental models. Nevertheless, the establishment of ES cell lines is difficult and the extensive use of PDX raises economic/ethical concerns. There is a growing consensus regarding the use of 3D cell culture to recapitulate physiological and pathophysiological features of human tissues, including drug sensitivity. Herein, we implemented a 3D cell culture methodology based on encapsulation of PDX-derived ES cell spheroids in alginate and maintenance in agitation-based culture systems. Under these conditions, ES cells displayed high proliferative and metabolic activity, while retaining the typical EWSR1-FLI1 chromosomal translocation. Importantly, 3D cultures presented reduced mouse PDX cell contamination compared to 2D cultures. Finally, we show that these 3D cultures can be employed in drug sensitivity assays, with results similar to those reported for the PDX of origin. In conclusion, this novel 3D cell culture method involving ES-PDX-derived cells is a suitable model to study ES pathobiology and can assist in the development of novel drugs against this disease, complementing PDX studies.

8.
J Control Release ; 324: 440-449, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32497782

ABSTRACT

Ewing sarcoma is a bone and soft tissue tumor predominantly affecting adolescents and young adults. To characterize changes in anticancer drug activity and intratumor drug distribution during the evolution of Ewing sarcomas, we used immunodeficient mice to establish pairs of patient-derived xenografts (PDX) at early (initial diagnosis) and late (relapse or refractory progression) stages of the disease from three patients. Analysis of copy number alterations (CNA) in early passage PDX tissues showed that two tumor pairs established from patients which responded initially to therapy and relapsed more than one year later displayed similar CNAs at early and late stages. For these two patients, PDX established from late tumors were more resistant to chemotherapy (irinotecan) than early counterparts. In contrast, the tumor pair established at refractory progression showed highly dissimilar CNA profiles, and the pattern of response to chemotherapy was discordant with those of relapsed cases. In mice receiving irinotecan infusions, the level of SN-38 (active metabolite of irinotecan) in the intracellular tumor compartment was reduced in tumors at later stages compared to earlier tumors for those pairs bearing similar CNAs, suggesting that distribution of anticancer drug shifted toward the extracellular compartment during clonal tumor evolution. Overexpression of the drug transporter P-glycoprotein in late tumor was likely responsible for this shift in drug distribution in one of the cases.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Pharmaceutical Preparations , Sarcoma, Ewing , Adolescent , Animals , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Humans , Irinotecan , Mice , Sarcoma, Ewing/drug therapy
9.
ACS Appl Mater Interfaces ; 11(42): 38483-38496, 2019 Oct 23.
Article in English | MEDLINE | ID: mdl-31537060

ABSTRACT

In this work, we designed, characterized, and investigated the performance of hydrolyzed galactomannan (hGM)-based amphiphilic nanoparticles for selective intratumoral accumulation in pediatric patient-derived sarcomas. To create a self-assembly amphiphilic copolymer, the side chain of hGM was hydrophobized with poly(methyl methacrylate) (PMMA) by utilizing a graft free radical polymerization reaction. Different hGM and MMA weight feeding ratios were used to adjust the critical aggregation concentration and the size and size distribution of the nanoparticles. The ability to actively target glucose transporter-1 (GLUT-1) was studied by fluorescence confocal microscopy and imaging flow cytometry in vitro on Rh30 (rhabdomyosarcoma) and patient-derived Ewing sarcoma (HSJD-ES-001) cell lines with different expression levels of GLUT-1. Results confirmed that the nanoparticles are internalized by ∼100% of the cells at 37 °C. Furthermore, we investigated the biodistribution of the nanoparticles in pediatric patient-derived models of two deadly musculoskeletal tumors, rhabdomyosarcoma and Ewing sarcoma. Outstandingly, the intratumoral accumulation of the nanoparticles correlated very well with the expression level of GLUT1 gene in each patient-derived tumor (P = 0.0141; Pearson's correlation test). Finally, we demonstrated the encapsulation capacity of these nanoparticles by loading 7.5% (w/w) of the hydrophobic first-generation tyrosine kinase inhibitor imatinib. These findings point out the potential of this new type of nanoparticle to target GLUT-1-expressing tumors and selectively deliver anticancer agents.


Subject(s)
Glucose Transporter Type 1/metabolism , Mannans/chemistry , Nanoparticles/chemistry , Animals , Cell Line, Tumor , Cell Survival/drug effects , Child , Drug Carriers/chemistry , Galactose/analogs & derivatives , Glucose Transporter Type 1/genetics , Heterografts , Humans , Imatinib Mesylate/chemistry , Imatinib Mesylate/therapeutic use , Mice , Microscopy, Confocal , Nanoparticles/metabolism , Nanoparticles/toxicity , Polymethyl Methacrylate/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/pathology , Tissue Distribution
10.
Sci Transl Med ; 11(476)2019 01 23.
Article in English | MEDLINE | ID: mdl-30674657

ABSTRACT

Retinoblastoma is a pediatric solid tumor of the retina activated upon homozygous inactivation of the tumor suppressor RB1 VCN-01 is an oncolytic adenovirus designed to replicate selectively in tumor cells with high abundance of free E2F-1, a consequence of a dysfunctional RB1 pathway. Thus, we reasoned that VCN-01 could provide targeted therapeutic activity against even chemoresistant retinoblastoma. In vitro, VCN-01 effectively killed patient-derived retinoblastoma models. In mice, intravitreous administration of VCN-01 in retinoblastoma xenografts induced tumor necrosis, improved ocular survival compared with standard-of-care chemotherapy, and prevented micrometastatic dissemination into the brain. In juvenile immunocompetent rabbits, VCN-01 did not replicate in retinas, induced minor local side effects, and only leaked slightly and for a short time into the blood. Initial phase 1 data in patients showed the feasibility of the administration of intravitreous VCN-01 and resulted in antitumor activity in retinoblastoma vitreous seeds and evidence of viral replication markers in tumor cells. The treatment caused local vitreous inflammation but no systemic complications. Thus, oncolytic adenoviruses targeting RB1 might provide a tumor-selective and chemotherapy-independent treatment option for retinoblastoma.


Subject(s)
Adenoviridae/physiology , Molecular Targeted Therapy , Oncolytic Viruses/physiology , Retinoblastoma Protein/metabolism , Retinoblastoma/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Mice , Neoplasm Metastasis , Rabbits , Retinoblastoma/immunology , Retinoblastoma/pathology , Survival Analysis , Tissue Distribution , Translational Research, Biomedical , Treatment Outcome , Virus Replication , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 25(7): 2228-2240, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30420447

ABSTRACT

PURPOSE: Endoglin (ENG; CD105) is a coreceptor of the TGFß family that is highly expressed in proliferating endothelial cells. Often coopted by cancer cells, ENG can lead to neo-angiogenesis and vasculogenic mimicry in aggressive malignancies. It exists both as a transmembrane cell surface protein, where it primarily interacts with TGFß, and as a soluble matricellular protein (sENG) when cleaved by matrix metalloproteinase 14 (MMP14). High ENG expression has been associated with poor prognosis in Ewing sarcoma, an aggressive bone cancer that primarily occurs in adolescents and young adults. However, the therapeutic value of ENG targeting has not been fully explored in this disease. EXPERIMENTAL DESIGN: We characterized the expression pattern of transmembrane ENG, sENG, and MMP14 in preclinical and clinical samples. Subsequently, the antineoplastic potential of two novel ENG-targeting monoclonal antibody-drug conjugates (ADC), OMTX503 and OMTX703, which differed only by their drug payload (nigrin-b A chain and cytolysin, respectively), was assessed in cell lines and preclinical animal models of Ewing sarcoma. RESULTS: Both ADCs suppressed cell proliferation in proportion to the endogenous levels of ENG observed in vitro. Moreover, the ADCs significantly delayed tumor growth in Ewing sarcoma cell line-derived xenografts and patient-derived xenografts in a dose-dependent manner. CONCLUSIONS: Taken together, these studies demonstrate potent preclinical activity of first-in-class anti-ENG ADCs as a nascent strategy to eradicate Ewing sarcoma.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Bone Neoplasms/metabolism , Endoglin/antagonists & inhibitors , Immunoconjugates/pharmacology , Sarcoma, Ewing/metabolism , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gene Expression , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Mice , Molecular Targeted Therapy , Precision Medicine , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/pathology , Xenograft Model Antitumor Assays
12.
Adv Healthc Mater ; 7(15): e1800255, 2018 08.
Article in English | MEDLINE | ID: mdl-29892999

ABSTRACT

Delivery of chemotherapy in the surgical bed has shown preclinical activity to control cancer progression upon subtotal resection of pediatric solid tumors, but whether this new treatment is safe for tumor-adjacent healthy tissues remains unknown. Here, Wistar rats are used to study the anatomic and functional impact of electrospun nanofiber matrices eluting SN-38-a potent chemotherapeutic agent-on several body sites where pediatric tumors such as neuroblastoma, Ewing sarcoma, and rhabdomyosarcoma arise. Blank and SN-38-loaded matrices embracing the femoral neurovascular bundle or in direct contact with abdominal viscera (liver, kidney, urinary bladder, intestine, and uterus) are placed. Foreign body tissue reaction to the implants is observed though no histologic damage in any tissue/organ. Skin healing is normal. Tissue reaction is similar for SN-38-loaded and blank matrices, with the exception of the hepatic capsule that is thicker for the former although within the limits consistent with mild foreign body reaction. Tissue and organ function is completely conserved after local treatments, as assessed by the rotarod test (forelimb function), hematologic tests (liver and renal function), and control of clinical signs. Overall, these findings support the clinical translation of SN-38-loaded nanofiber matrices to improve local control strategies of surgically resected tumors.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Irinotecan/chemistry , Nanofibers/chemistry , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Mice , Rats, Wistar , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/metabolism
13.
J Control Release ; 276: 59-71, 2018 04 28.
Article in English | MEDLINE | ID: mdl-29501533

ABSTRACT

We report for the first time on a nano-drug delivery system based on glucosylated polymeric nanomicelles to actively target the second-generation tyrosine kinase inhibitor dasatinib to glucose-avid pediatric sarcomas by the intravenous route. After a comprehensive physicochemical characterization that confirmed the substantially lower critical micellar concentration and the higher encapsulation capacity of the glucosylated amphiphilic nanocarrier with respect to the pristine counterpart, we showed a 9-fold decrease of the half maximal inhibitory concentration of dasatinib in a rhabdomyosarcoma cell line, Rh30, in vitro. In immunodeficient mice bearing the glucose-avid Rh30 xenograft, we revealed that the glucosylated polymeric nanomicelles increased the delivery of dasatinib in the tumor parenchyma. Conversely, the exposure of off-target tissues and organs to the drug was substantially reduced. Upon experimental confirmation that most patient-derived xenograft (PDX) models of pediatric sarcomas overexpress glucose transporter 1 (GLUT-1), we demonstrated the selective accumulation of dasatinib in a patient-derived rhabdomyosarcoma model in vivo. Conversely, the reference dose administered by the oral route was not tumor-selective. Finally, the improved nanocarrier pharmacokinetics led to prolonged median survival of mice bearing a clinically relevant PDX model of alveolar rhabdomyosarcoma from 19 days for the untreated controls to 27 days for the targeted therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Dasatinib/administration & dosage , Micelles , Nanostructures/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Child , Dasatinib/pharmacokinetics , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Mice, Nude , Protein Kinase Inhibitors/pharmacokinetics , Sarcoma/drug therapy , Sarcoma/genetics , Sarcoma/metabolism
14.
J Control Release ; 264: 34-44, 2017 Oct 28.
Article in English | MEDLINE | ID: mdl-28830790

ABSTRACT

Treatment of retinoblastoma -a pediatric cancer of the developing retina- might benefit from strategies to inhibit the blood-retinal barrier (BRB). The potent anticancer agent topotecan is a substrate of efflux transporters BCRP and P-gp, which are expressed at the BRB to restrict vitreous and retinal distribution of xenobiotics. In this work we have studied vitreous and retinal distribution, tumor accumulation and antitumor activity of topotecan, using pantoprazole as inhibitor of BCRP and P-gp. We used rabbit and mouse eyes as BRB models and patient-derived xenografts as retinoblastoma models. To validate the rabbit BRB model we stained BCRP and P-gp in the retinal vessels. Using intravitreous microdialysis we showed that the penetration of the rabbit vitreous by lactone topotecan increased significantly upon concomitant administration of pantoprazole (P=0.0285). Pantoprazole also increased topotecan penetration of the mouse vitreous, measured as the vitreous-to-plasma topotecan concentration ratio at the steady state (P=0.0246). Pantoprazole increased topotecan antitumor efficacy and intracellular penetration in retinoblastoma in vitro, but did not enhance intratumor drug distribution and survival in mice bearing the intraocular human tumor HSJD-RBT-2. Anatomical differences with the clinical setting likely limited our in vivo study, since xenografts were poorly vascularized masses that loaded most of the vitreous compartment. We conclude that pharmacological modulation of the BRB is feasible, enhances anticancer drug distribution into the vitreous and might have clinical implications in retinoblastoma. CHEMICAL COMPOUNDS INCLUDED IN THIS MANUSCRIPT: Topotecan (PubChem CID: 60700) Pantoprazole sodium (PubChem CID: 15008962).


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/pharmacology , Blood-Retinal Barrier/drug effects , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Topoisomerase I Inhibitors/therapeutic use , Topotecan/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Blood-Retinal Barrier/metabolism , Humans , Mice, Nude , Pantoprazole , Rabbits , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinoblastoma/genetics , Retinoblastoma/metabolism , Topoisomerase I Inhibitors/pharmacokinetics , Topotecan/pharmacokinetics , Vitreous Body/metabolism , Xenograft Model Antitumor Assays
15.
J Control Release ; 255: 108-119, 2017 06 10.
Article in English | MEDLINE | ID: mdl-28412222

ABSTRACT

Neuroblastoma is a pediatric solid tumor with high expression of the tumor associated antigen disialoganglioside GD2. Despite initial response to induction therapy, nearly 50% of high-risk neuroblastomas recur because of chemoresistance. Here we encapsulated the topoisomerase-I inhibitor SN-38 in polymeric nanoparticles (NPs) surface-decorated with the anti-GD2 mouse mAb 3F8 at a mean density of seven antibody molecules per NP. The accumulation of drug-loaded NPs targeted with 3F8 versus with control antibody was monitored by microdialysis in patient-derived GD2-expressing neuroblastoma xenografts. We showed that the extent of tumor penetration by SN-38 was significantly higher in mice receiving the targeted nano-drug delivery system when compared to non-targeted system or free drug. This selective penetration of the tumor extracellular fluid translated into a strong anti-tumor effect prolonging survival of mice bearing GD2-high neuroblastomas in vivo.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/analogs & derivatives , Extracellular Fluid/metabolism , Immunoglobulin G/administration & dosage , N-Acetylgalactosaminyltransferases/antagonists & inhibitors , Nanoparticles/administration & dosage , Neuroblastoma/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Murine-Derived , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/chemistry , Camptothecin/pharmacokinetics , Cell Line, Tumor , Child, Preschool , Drug Liberation , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulin G/chemistry , Irinotecan , Male , Mice, Nude , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/immunology , N-Acetylgalactosaminyltransferases/metabolism , Nanoparticles/chemistry , Neuroblastoma/drug therapy , Tissue Distribution , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...