Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Blood Rev ; 64: 101164, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38216442

ABSTRACT

Hemophilia A, an X-linked genetic disorder, is characterized by a deficiency or dysfunction of clotting Factor VIII. The treatment landscape has substantially changed by introducing novel extended half-life factor VIII (EHL-FVIII) replacement therapies such as efanesoctocog Alfa and non-factor replacement therapy such as emicizumab. These agents signal a shift from treatments requiring multiple weekly infusions to advanced therapies with long half-lives, offering superior protection against bleeding and improving patient adherence and quality of life. While EHL-FVIII treatment might lead to inhibitor development in some patients, non-factor replacement therapy carries thrombotic risks. Therefore, ongoing research and the generation of robust clinical evidence remain vital to guide the selection of optimal and cost-effective first-line therapies for hemophilia A patients.


Subject(s)
Hemophilia A , Hemostatics , Humans , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Half-Life , Quality of Life , Hemostatics/therapeutic use
2.
Pediatrics ; 149(6)2022 06 01.
Article in English | MEDLINE | ID: mdl-35543028

ABSTRACT

Although rare in pediatrics, ischemic priapism carries significant risk of erectile dysfunction if not treated promptly. We report a case of idiopathic ischemic priapism in a male aged 14 years, refractory to traditional therapies, including aspiration/irrigation, phenylephrine injection, and distal shunt. However, the priapism resolved after intracavernosal injections of tissue plasminogen activator (tPA) with preservation of normal erectile function. To our knowledge, this is the first reported case of intracavernosal tPA for treatment of pediatric priapism. tPA may be a useful management option for recalcitrant ischemic priapism in pediatric patients and may prevent devastating outcomes such as lifelong erectile dysfunction.


Subject(s)
Erectile Dysfunction , Pediatrics , Priapism , Child , Humans , Ischemia/complications , Male , Penis , Priapism/drug therapy , Priapism/etiology , Tissue Plasminogen Activator/therapeutic use
4.
Mol Imaging Biol ; 22(2): 434-443, 2020 04.
Article in English | MEDLINE | ID: mdl-31183841

ABSTRACT

PURPOSE: There is a continuous search for imaging techniques with high sensitivity and specificity for brain tumors. Positron emission tomography (PET) imaging has shown promise, though many PET agents either have a low tumor specificity or impractical physical half-lives. [124I]CLR1404 is a small molecule alkylphosphocholine analogue that is thought to bind to plasma membrane lipid rafts and has shown high tumor-to-background ratios (TBR) in a previous pilot study in brain tumor patients. This study attempts to define the clinical value of [124I]CLR1404 PET/CT (aka CLR124). PROCEDURES: Adult patients with new or suspected recurrence of high-grade primary or metastatic brain tumors (N = 27) were injected with [124I]CLR1404 followed by PET/CT at 6, 24, and 48 h. Standard uptake values (SUV) and TBR values were calculated for all time points. Uptake of [124I]CLR1404 was qualitatively assessed, compared with magnetic resonance imaging (MRI), and correlated with clinical outcome. Final diagnosis (N = 25) was established based on surgically resected tissue or long-term follow-up. RESULTS: Positive uptake with high TBR was detected in all but one patient with a final diagnosis of primary/recurrent brain tumor (12/13) and in less than half of patients with treatment-related changes (5/12). Concordance between [124I]CLR1404 uptake and contrast enhancement on MRI was seen in < 40 %, with no concordance between T2-hyperintensities and uptake. No significant difference in overall outcome was found between patients with and without [124I]CLR1404 uptake. CONCLUSIONS: The uptake pattern in these patients suggests a very high sensitivity of [124I]CLR1404 PET/CT for diagnosing tumor tissue; however, tumor specificity needs to be further defined. Relative lack of concordance with standard MRI characteristics suggests that [124I]CLR1404 PET/CT provides additional information about brain tumors compared to MRI alone, potentially improving clinical decision-making.


Subject(s)
Brain Neoplasms/diagnostic imaging , Iodine Radioisotopes , Iodobenzenes , Membrane Microdomains/chemistry , Neoplasm Metastasis , Phospholipid Ethers , Positron Emission Tomography Computed Tomography , Adult , Aged , Brain/diagnostic imaging , Brain Neoplasms/pathology , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/secondary , Decision Making , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Predictive Value of Tests , Prognosis
5.
J Nucl Med ; 60(10): 1414-1420, 2019 10.
Article in English | MEDLINE | ID: mdl-30926646

ABSTRACT

Cancer is the second leading cause of death for children between the ages of 5 and 14 y. For children diagnosed with metastatic or recurrent solid tumors, for which the utility of external-beam radiotherapy is limited, the prognosis is particularly poor. The availability of tumor-targeting radiopharmaceuticals for molecular radiotherapy (MRT) has demonstrated improved outcomes in these patient populations, but options are nonexistent or limited for most pediatric solid tumors. 18-(p-iodophenyl)octadecylphosphocholine (CLR1404) is a novel antitumor alkyl phospholipid ether analog that broadly targets cancer cells. In this study, we evaluated the in vivo pharmacokinetics of 124I-CLR1404 (CLR 124) and estimated theranostic dosimetry for 131I-CLR1404 (CLR 131) MRT in murine xenograft models of the pediatric solid tumors neuroblastoma, rhabdomyosarcoma, and Ewing sarcoma. Methods: Tumor-bearing mice were imaged with small-animal PET/CT to evaluate the whole-body distribution of CLR 124 and, correcting for differences in radioactive decay, predict that of CLR 131. Image volumes representing CLR 131 provided input for Geant4 Monte Carlo simulations to calculate subject-specific tumor dosimetry for CLR 131 MRT. Pharmacokinetics for CLR 131 were extrapolated to adult and pediatric humans to estimate normal-tissue dosimetry. In neuroblastoma, a direct comparison of CLR 124 with 124I-metaiodobenzylguanidine (124I-MIBG) in an MIBG-avid model was performed. Results: In vivo pharmacokinetics of CLR 124 showed selective uptake and prolonged retention across all pediatric solid tumor models investigated. Subject-specific tumor dosimetry for CLR 131 MRT presents a correlative relationship with tumor-growth delay after CLR 131 MRT. Peak uptake of CLR 124 was, on average, 22% higher than that of 124I-MIBG in an MIBG-avid neuroblastoma model. Conclusion: CLR1404 is a suitable theranostic scaffold for dosimetry and therapy with potentially broad applicability in pediatric oncology. Given the ongoing clinical trials for CLR 131 in adults, these data support the development of pediatric clinical trials and provide detailed dosimetry that may lead to improved MRT treatment planning.


Subject(s)
Iodine Radioisotopes/pharmacology , Neoplasms/diagnostic imaging , Neoplasms/therapy , 3-Iodobenzylguanidine/pharmacology , Animals , Cell Line, Tumor , Child , Computer Simulation , Disease Models, Animal , Humans , Iodobenzenes/pharmacology , Mice , Mice, Inbred NOD , Monte Carlo Method , Neoplasm Recurrence, Local , Neoplasm Transplantation , Phospholipid Ethers/pharmacology , Positron Emission Tomography Computed Tomography , Prognosis , Radiometry , Radiopharmaceuticals , Theranostic Nanomedicine
6.
Leuk Lymphoma ; 60(3): 598-609, 2019 03.
Article in English | MEDLINE | ID: mdl-30187806

ABSTRACT

Allogeneic hematopoietic stem cell transplantation is a curative option for patients with a variety of diseases. Transplantation from a related haploidentical donor is being increasingly utilized for patients who lack an available human leukocyte antigen matched related or unrelated donor. One of the strategies used for haploidentical transplants involves selective depletion of T cells expressing the αß T cell receptor and CD19+ B cells prior to transplant. This allows for the removal of cells responsible for graft-versus-host disease and post-transplant lymphoproliferative disorder but maintains hematopoietic progenitor and stem cells for engraftment (CD34+ cells), as well as cells to elicit graft-versus-tumor effect and provide anti-infective activity (such as gamma-delta T cells and natural killer cells). The aim of this review article is to present and discuss the data available to date from studies utilizing this method of transplantation.


Subject(s)
Antigens, CD19/metabolism , B-Lymphocytes/metabolism , Hematopoietic Stem Cell Transplantation , Lymphocyte Depletion , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Transplantation, Haploidentical , B-Lymphocytes/immunology , Graft Rejection/immunology , Graft Survival , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Immune Reconstitution , Lymphocyte Depletion/methods , Mortality , Opportunistic Infections/etiology , Transplantation, Haploidentical/adverse effects , Transplantation, Haploidentical/methods , Transplantation, Homologous , Treatment Outcome
7.
Mol Cancer Ther ; 17(11): 2320-2328, 2018 11.
Article in English | MEDLINE | ID: mdl-30108133

ABSTRACT

Antitumor alkyl phospholipid (APL) analogs comprise a group of structurally related molecules with remarkable tumor selectivity. Some of these compounds have shown radiosensitizing capabilities. CLR127 is a novel, clinical-grade antitumor APL ether analog, a subtype of synthetic APL broadly targeting cancer cells with limited uptake in normal tissues. The purpose of this study was to investigate the effect of CLR127 to modulate radiation response across several adult and pediatric cancer types in vitro as well as in murine xenograft models of human prostate adenocarcinoma, neuroblastoma, Ewing sarcoma, and rhabdomyosarcoma. In vitro, CLR127 demonstrated selective uptake in cancer cells compared to normal cells. In cancer cells, CLR127 treatment prior to radiation significantly decreased clonogenic survival in vitro, and led to increased radiation-induced double-stranded DNA (dsDNA) breakage compared with radiation alone, which was not observed in normal controls. In animal models, CLR127 effectively increased the antitumor response to fractionated radiotherapy and led to delayed tumor regrowth at potentially clinically achievable doses. In conclusion, our study highlights the ability of CLR127 to increase radiation response in several cancer types. Given almost universal uptake of CLR127 in malignant cells, future research should test whether the observed effects can be extended to other tumor types. Our data provide a strong rationale for clinical testing of CLR127 as a tumor-targeted radiosensitizing agent. Mol Cancer Ther; 17(11); 2320-8. ©2018 AACR.


Subject(s)
Neoplasms/pathology , Phospholipid Ethers/pharmacology , Radiation Tolerance , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells , DNA Damage , Histones/metabolism , Humans , Mice, Nude , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , X-Rays , Xenograft Model Antitumor Assays
8.
Health Phys ; 114(4): 450-459, 2018 04.
Article in English | MEDLINE | ID: mdl-29481536

ABSTRACT

There is a growing need to estimate the absorbed dose to small animals from preclinical investigations involving diagnostic and therapeutic radiopharmaceuticals. This paper introduces a Monte Carlo-based dosimetry platform called RAPID, which is capable of calculating murine-specific three-dimensional (3D) dose distributions. A comparison is performed between absorbed doses calculated with RAPID and absorbed doses calculated in a commonly used reference mouse phantom called MOBY. Four test mice containing different xenografts underwent serial PET/CT imaging using a novel diagnostic therapy (theranostic) agent NM404, which can be labeled with I for imaging or I for therapy. Using the PET/CT data, 3D dose distributions from I-NM404 were calculated in the mice using RAPID. Mean organ doses in these four test mice were compared to mean organ doses derived by using two previously published I S-values datasets in MOBY. In addition, mean tumor doses calculated in RAPID were compared to mean organ doses derived from unit density spheres. Large differences were identified between mean organ doses calculated in the test mice using RAPID and those derived in the MOBY phantom. Mean absorbed dose percent errors in organs ranged between 0.3% and 333%. Overall, mass scaling improved agreement between MOBY phantom calculations and RAPID, where percent errors were all less than 26%, with the exception of the lung in which percent errors reached values of 48%. Percent errors in mean tumor doses in the test mice and unit density spheres were less pronounced but still ranged between 8% and 23%. This work demonstrates the limitations of using pre-computed S-values in computational phantoms to predict organ doses in small animals from theranostic procedures. RAPID can generate accurate 3D dose distributions in small animals and in turn offer much greater insight on the ability of a given theranostic agent to image and treat diseases.


Subject(s)
Drug Evaluation, Preclinical , Iodine Radioisotopes/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Phantoms, Imaging , Animals , Computer Simulation , Humans , Iodine Radioisotopes/administration & dosage , Male , Mice , Mice, Inbred NOD , Monte Carlo Method , Neoplasms/pathology , Positron Emission Tomography Computed Tomography/methods , Radiation Dosage , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
J Nucl Med ; 59(2): 244-250, 2018 02.
Article in English | MEDLINE | ID: mdl-28747518

ABSTRACT

External-beam radiotherapy plays a critical role in the treatment of most pediatric solid tumors. Particularly in children, achieving an optimal therapeutic index to avoid damage to normal tissue is extremely important. Consequently, in metastatic disease, the utility of external-beam radiotherapy is limited. Molecular radiotherapy with tumor-targeted radionuclides may overcome some of these challenges, but to date there exists no single cancer-selective agent capable of treating various pediatric malignancies independently of their histopathologic origin. We tested the therapeutic potential of the clinical-grade alkyl-phospholipid ether analog CLR1404, 18-(p-iodophenyl)octadecyl phosphocholine, as a scaffold for tumor-targeted radiotherapy of pediatric malignancies. Methods: Uptake of CLR1404 by pediatric solid tumor cells was tested in vitro by flow cytometry and in vivo by PET/CT imaging and dosimetry. The therapeutic potential of 131I-CLR1404 was evaluated in xenograft models. Results: In vitro, fluorescent CLR1404-BODIPY showed significant selective uptake in a variety of pediatric cancer lines compared with normal controls. In vivo tumor-targeted uptake in mouse xenograft models using 124I-CLR1404 was confirmed by imaging. Single-dose intravenous injection of 131I-CLR1404 significantly delayed tumor growth in all rodent pediatric xenograft models and extended animal survival while demonstrating a favorable side effect profile. Conclusion:131I-CLR1404 has the potential to become a tumor-targeted radiotherapeutic drug with broad applicability in pediatric oncology. Because 131I-CLR1404 has entered clinical trials in adults, our data warrant the development of pediatric clinical trials for this particularly vulnerable patient population.


Subject(s)
Iodobenzenes/chemistry , Iodobenzenes/therapeutic use , Neoplasms/radiotherapy , Phospholipid Ethers/chemistry , Phospholipid Ethers/therapeutic use , Alkylation , Animals , Biological Transport , Cell Line, Tumor , Cell Transformation, Neoplastic , Child , Humans , Iodobenzenes/metabolism , Mice , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/pathology , Phospholipid Ethers/metabolism , Positron Emission Tomography Computed Tomography , Survival Analysis
10.
J Mater Chem B ; 4(3): 513-520, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-27087966

ABSTRACT

Although nanomaterials have been widely investigated for drug delivery, imaging and immunotherapy, their potential roles in triggering innate cellular immune responses while simultaneously serving as imaging enhancer remain unexplored. In this work, gold nanoparticles (GNPs) conjugated to the tumor-targeting anti-GD2 antibody hu14.18K322A, namely HGNPs, were designed and synthesized to specifically enhance computerized tomography (CT) imaging contrast and to stimulate the attack of neuroblastoma and melanoma cells by natural killer (NK) cells. The HGNPs specifically targeted GD2-positive neuroblastoma (NB1691) and melanoma (M21) cells, with an enhancement of CT contrast images of the HGNP-labeled cell pellets by 5.27- and 7.66-fold, respectively, compared to images of unlabeled cell pellets. The HGNPs also triggered NK-mediated antibody-dependent cellular cytotoxicity (ADCC) in NB1691 and M21 cells with a two-fold higher efficacy compared to that elicited by hu14.18K322A alone, with no adverse effect to GD2-negative PC-3 cells. These results suggest that HGNPs are promising theranostic agents for neuroblastoma and melanoma cancers.

11.
Nanomedicine (Lond) ; 10(19): 2973-2988, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26420448

ABSTRACT

AIM: To develop biocompatible, tumor-specific multifunctional iron-oxide nanoconstructs targeting neuroblastoma, an aggressive pediatric malignancy. MATERIALS & METHODS: Clinical-grade humanized monoclonal antibody (hu14.18K322A), designed to target GD2 antigen on neuroblastoma with reduced nonspecific immune interactions, was conjugated to hydroxyethyl starch-coated iron-oxide nanoparticles. Targeting capability in vitro and in vivo was assessed by immunofluorescence, electron microscopy, analytical spectrophotometry, histochemistry and magnetic resonance R2* relaxometry. RESULTS: The biocompatible nanoconstructs demonstrated high tumor specificity in vitro and in vivo, and low background uptake in a mouse flank xenograft model. Specific accumulation in tumors enabled particle visualization and quantification by magnetic resonance R2* mapping. CONCLUSION: Our findings support the further development toward clinical application of this anti-GD2 iron-oxide nanoconstruct as diagnostic and therapeutic scaffold for neuroblastoma and potentially other GD2-positive malignancies.

12.
J Pediatr Hematol Oncol ; 37(2): 79-93, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25590232

ABSTRACT

The past decade has seen several anticancer immunotherapeutic strategies transition from "promising preclinical models" to treatments with proven clinical activity or benefit. In 2013, the journal Science selected the field of Cancer Immunotherapy as the overall number-1 breakthrough for the year in all of scientific research. In the setting of cancer immunotherapy for adult malignancies, many of these immunotherapy strategies have relied on the cancer patient's endogenous antitumor T-cell response. Although much promising research in pediatric oncology is similarly focused on T-cell reactivity, several pediatric malignancies themselves, or the chemo-radiotherapy used to achieve initial responses, can be associated with profound immune suppression, particularly of the T-cell system. A separate component of the immune system, also able to mediate antitumor effects and less suppressed by conventional cancer treatment, is the NK-cell system. In recent years, several distinct immunotherapeutic approaches that rely on the activity of NK cells have moved from preclinical development into clinical testing, and some have shown clear antitumor benefit. This review provides an overview of NK cell-based immunotherapy efforts that are directed toward childhood malignancies, with an emphasis on protocols that are already in clinical testing.


Subject(s)
Immunotherapy , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , Adult , Child , Humans , Prognosis
13.
Am J Cancer Res ; 5(11): 3422-35, 2015.
Article in English | MEDLINE | ID: mdl-26807322

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid tumor in children and is associated with high mortality in advanced stages. Survivors suffer from long-term treatment-related sequelae. Thus, new targeted treatment options are urgently needed. 18-(p-[(127)I] iodophenyl) octadecyl phosphocholine (CLR1404) is a novel, broadly tumor targeted small molecule drug suitable for intravenous injection with highly selective tumor uptake. As a carrier molecule for radioactive iodine, CLR1404 is in clinical trials as cancer imaging agent and radiotherapeutic drug. Chemically, CLR1404 belongs to the anti-tumor alkyl phospholipids, a class of drugs known to have intrinsic cytotoxic effects on cancer cells. Therefore, we hypothesized that CLR1404 could be a tumor-targeted anti-cancer agent for neuroblastoma, and investigated its effect in vitro and in vivo. CLR1404 was taken up by NB cells in a highly tumor-selective manner both in vitro and in vivo, confirmed by flow cytometry and PET/CT imaging of mouse flank xenografts with (124)I-CLR1404, respectively. Using flow cytometry, MTT assay, Western blotting and caspase 3/7 assay, we confirm that in vitro treatment with CLR1404 leads to robust apoptosis and cell death in multiple NB cell lines and is associated with Akt inhibition, while sparing normal cells. Treatment with CLR1404 in doses of 10 or 30 mg/kg administered by intravenous injection once weekly for 7 weeks significantly inhibited the tumor growth rate in a mouse flank xenograft model of NB (P<0.001) when compared to control cohorts, without causing drug-related hematotoxicity or other noticeable adverse effects, which was determined by serial tumor volume measurements, complete blood counts, and monitoring of animal-specific health parameters. We conclude that CLR1404 warrants clinical exploration as a novel, tumor selective anticancer agent in NB and potentially other cancers.

14.
Future Oncol ; 10(9): 1659-78, 2014.
Article in English | MEDLINE | ID: mdl-25145434

ABSTRACT

Novel immune-based therapies are becoming available as additions to, and in some cases as alternatives to, the traditional treatment modalities such as chemotherapy, surgery and radiation that have improved outcomes for childhood cancer for decades. In this article, we will discuss what immunotherapies are being tested in the clinic, barriers to widespread application, and the future of immuno-oncology for childhood cancer. While in many cases, these therapies have shown dramatic responses in the setting of refractory or relapsed cancer, much remains to be learned about how to integrate these therapies into existing upfront regimens. The progress and challenges of developing immunotherapies for childhood cancer in a timely and cost-effective fashion will be discussed.


Subject(s)
Immunotherapy , Neoplasms/therapy , Cancer Vaccines , Child , Humans , Neoplasms/immunology , Pediatrics
15.
J Clin Oncol ; 32(14): 1445-52, 2014 May 10.
Article in English | MEDLINE | ID: mdl-24711551

ABSTRACT

PURPOSE: The addition of immunotherapy, including a combination of anti-GD2 monoclonal antibody (mAb), ch14.18, and cytokines, improves outcome for patients with high-risk neuroblastoma. However, this therapy is limited by ch14.18-related toxicities that may be partially mediated by complement activation. We report the results of a phase I trial to determine the maximum-tolerated dose (MTD), safety profile, and pharmacokinetics of hu14.18K322A, a humanized anti-GD2 mAb with a single point mutation (K322A) that reduces complement-dependent lysis. PATIENTS AND METHODS: Eligible patients with refractory or recurrent neuroblastoma received escalating doses of hu14.18K322A ranging from 2 to 70 mg/m(2) per day for 4 consecutive days every 28 days (one course). RESULTS: Thirty-eight patients (23 males; median age, 7.2 years) received a median of two courses (range, one to 15). Dose-limiting grade 3 or 4 toxicities occurred in four of 36 evaluable patients and were characterized by cough, asthenia, sensory neuropathy, anorexia, serum sickness, and hypertensive encephalopathy. The most common non-dose-limiting grade 3 or 4 toxicities during course one were pain (68%) and fever (21%). Six of 31 patients evaluable for response by iodine-123 metaiodobenzylguanidine score had objective responses (four complete responses; two partial responses). The first-course pharmacokinetics of hu14.18K322A were best described by a two-compartment linear model. Median hu14.18K322A α (initial phase) and ß (terminal phase) half-lives were 1.74 and 21.1 days, respectively. CONCLUSION: The MTD, and recommended phase II dose, of hu14.18K322A is 60 mg/m(2) per day for 4 days. Adverse effects, predominately pain, were manageable and improved with subsequent courses.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Brain Neoplasms/drug therapy , Neuroblastoma/drug therapy , Adolescent , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Child , Child, Preschool , Female , Humans , Male , Maximum Tolerated Dose , Recurrence
16.
J Mater Chem B ; 2(37): 6198-6206, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-26660881

ABSTRACT

Specific targeting is a key step to realize the full potential of iron oxide nanoparticles in biomedical applications, especially tumor-associated diagnosis and therapy. Here, we developed anti-GD2 antibody conjugated iron oxide nanoparticles for highly efficient neuroblastoma cell targeting. The antibody conjugation was achieved through an easy, linker-free method based on catechol reactions. The targeting efficiency and specificity of the antibody-conjugated nanoparticles to GD2-positive neuroblastoma cells were confirmed by flow cytometry, fluorescence microscopy, Prussian blue staining and transmission electron microscopy. These detailed studies indicated that the receptor-recognition capability of the antibody was fully retained after conjugation and the conjugated nanoparticles quickly attached to GD2-positive cells within four hours. Interestingly, longer treatment (12 h) led the cell membrane-bound nanoparticles to be internalized into cytosol, either by directly penetrating the cell membrane or escaping from the endosomes. Last but importantly, the uniquely designed functional surfaces of the nanoparticles allow easy conjugation of other bioactive molecules.

17.
Curr Pharm Des ; 19(37): 6606-21, 2013.
Article in English | MEDLINE | ID: mdl-23621531

ABSTRACT

Iron oxide (IO) nanoparticles hold great promise as diagnostic and therapeutic agents in oncology. Their intrinsic physical properties make IO nanoparticles particularly interesting for simultaneous drug delivery, molecular imaging, and applications such as localized hyperthermia. Multiple non-targeted IO nanoparticle preparations have entered clinical trials, but more exciting, new tumortargeted IO nanoparticle preparations are currently being tested in preclinical settings. This paper will analyze the challenges faced by this new theranostic modality, with a specific focus on the interactions of IO nanoparticles with the innate and adaptive immune systems, and their effect on nanoparticle biodistribution and tumor targeting. Next, we will review the critical need for innovative surface chemistry solutions and strategies to overcome the immune interactions that prevent existing tumor-targeted IO preparations from entering clinical trials. Finally, we will provide an outlook for the future role of IO nanoparticles in oncology, which have the promise of becoming significant contributors to improved diagnosis and treatment of cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Diagnostic Imaging , Ferric Compounds/chemistry , Immune System , Nanoparticles/administration & dosage , Neoplasms/diagnosis , Neoplasms/drug therapy , Animals , Humans , Nanoparticles/therapeutic use , Neoplasms/immunology
18.
J Pediatr Hematol Oncol ; 34(3): 232-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22082744

ABSTRACT

Inflammatory pseudotumors (IPTs) are rare, enigmatic lesions that may develop as a late manifestation of a reparative process. We describe the case of a teenager with primary mediastinal subtype of diffuse large B-cell lymphoma who developed an IPT at the site of the original lymphoma, mimicking relapse of disease on positron emission tomography/computed tomography imaging. This is the first report of IPT in a teenager with mediastinal lymphoma. This case is an important reminder of the limitations of positron emission tomography/computed tomography imaging in patients with lymphoma and stresses the importance of histologic confirmation of suspected treatment failure or relapse.


Subject(s)
Fluorodeoxyglucose F18 , Histiocytosis/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Mediastinal Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Positron-Emission Tomography , Radiopharmaceuticals , Xanthomatosis/pathology , Adolescent , Female , Histiocytosis/drug therapy , Histiocytosis/etiology , Humans , Lymphoma, Large B-Cell, Diffuse/complications , Lymphoma, Large B-Cell, Diffuse/drug therapy , Mediastinal Neoplasms/complications , Mediastinal Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/etiology , Prognosis , Xanthomatosis/drug therapy , Xanthomatosis/etiology
19.
J Am Chem Soc ; 133(35): 13918-21, 2011 Sep 07.
Article in English | MEDLINE | ID: mdl-21827210

ABSTRACT

To conquer complex and devastating diseases such as cancer, more coordinated and combined attack strategies are needed. We suggest that these can be beautifully achieved by using nanoconstruct design. We present an example showing that neuroblastoma cells are selectively killed by a nanoconstruct that specifically targets neuroblastoma cells, pushes cells to the vulnerable phase of the cell cycle, and greatly enhances radiation-induced cell death. The success of this multipronged attack approach launched by cell-embedded nanoconstructs demonstrates the power and flexibility of nanotechnology in treating cancer, a difficult task for a small molecule.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Delivery Systems/methods , Nanostructures/chemistry , Neuroblastoma/therapy , Paclitaxel/administration & dosage , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Humans , Nanomedicine/methods , Nanostructures/analysis , X-Rays
20.
Pain ; 149(1): 135-142, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20171010

ABSTRACT

Monoclonal antibodies against GD(2) ganglioside, such as ch14.18, the human-mouse chimeric antibody, have been shown to be effective for the treatment of neuroblastoma. However, treatment is associated with generalized, relatively opiate-resistant pain. We investigated if a point mutation in ch14.18 antibody (hu14.18K332A) to limit complement-dependent cytotoxicity (CDC) would ameliorate the pain behavior, while preserving antibody-dependent cellular cytotoxicity (ADCC). In vitro, CDC and ADCC were measured using europium-TDA assay. In vivo, allodynia was evaluated by measuring thresholds to von Frey filaments applied to the hindpaws after injection of either ch14.18 or hu14.18K332 into wild type rats or rats with deficient complement factor 6. Other rats were pretreated with complement factor C5a receptor antagonist and tested following ch14.18 injection. The mutation reduces the antibody's ability to activate complement, while maintaining its ADCC capabilities. Injection of hu14.18K322 (1 or 3mg/kg) produced faster resolving allodynia than that engendered by ch14.18 (1mg/kg). Injection of ch14.18 (1mg/kg) into rats with C6 complement deficiency further reduced antibody-induced allodynia, while pre-treatment with complement factor C5a receptor antagonist completely abolished ch14.18-induced allodynia. These findings showed that mutant hu14.18 K322 elicited less allodynia than ch14.18 and that ch14.18-elicited allodynia is due to activation of the complement cascade: in part, to formation of membrane attack complex, but more importantly to release of complement factor C5a. Development of immunotherapeutic agents with decreased complement-dependent lysis while maintaining cellular cytotoxicity may offer treatment options with reduced adverse side effects, thereby allowing dose escalation of therapeutic antibodies.


Subject(s)
Antibodies/genetics , Antibodies/immunology , Gangliosides/immunology , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Animals , Antibodies/adverse effects , Immunoglobulin Fc Fragments/adverse effects , Immunoglobulin Fc Fragments/genetics , Male , Point Mutation/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...