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










Publication year range
1.
Cardiovasc Intervent Radiol ; 47(5): 661-669, 2024 May.
Article in English | MEDLINE | ID: mdl-38609583

ABSTRACT

PURPOSE: Antiangiogenic agents have been used for many years as a first-line systemic treatment for advanced HCC. Embolization with cytostatic drugs on the other hand is the first-line treatment for intermediate HCC. The two types of drugs have not been combined for intraarterial delivery yet. The loading and release dynamics and the in vitro effect of their combination are tested in this experimental study. MATERIALS AND METHODS: Drug-eluting beads were loaded with doxorubicin, sunitinib and sunitinib analogue piperazine (SAP) alone and with their combinations. Diameter change, loading, release, and effect in cellular proliferation were assessed. RESULTS: The average microsphere diameter after loading was 473.7 µm (µm) for Doxorubicin, 388.4 µm for Sunitinib, 515.5 µm for SAP, 414.8 µm for the combination Doxorubicin/Sunitinib and 468.8 µm for the combination Doxorubicin /SAP. Drug release in 0.9% NaCl was 10% for Doxorubicin, 49% for Sunitinib, 25% for SAP, 20%/18% for the combination Doxorubicin/Sunitinib, and 18%/23% for the combination Doxorubicin/SAP whereas in human plasma it was 56%, 27%, 13%, 76%/63% and 62%/15%, respectively. The mean concentration of Doxorubicin that led to inhibition of 50% of cellular proliferation in an HCC Huh7 cell line was 163.1 nM (nM), for Sunitinib 10.3 micromolar (µΜ), for SAP 16.7 µΜ, for Doxorubicin/Sunitinib 222.4 nM and for Doxorubicin/SAP 275 nM. CONCLUSIONS: Doxorubicin may be combined with antiangiogenic drugs with satisfactory in vitro loading and release outcomes and effect on cellular lines.


Subject(s)
Angiogenesis Inhibitors , Carcinoma, Hepatocellular , Doxorubicin , Indoles , Liver Neoplasms , Sunitinib , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Doxorubicin/analogs & derivatives , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Sunitinib/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Angiogenesis Inhibitors/administration & dosage , Humans , Microspheres , Cell Proliferation/drug effects , Pyrroles/administration & dosage , Piperazines/therapeutic use , Cell Line, Tumor , Chemoembolization, Therapeutic/methods , In Vitro Techniques , Drug Liberation
2.
J Med Chem ; 65(1): 271-284, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34967607

ABSTRACT

c(RGDyK)-based conjugates of gemcitabine (GEM) with the carbonate and carbamate linkages in the 6-OH group of GEM were synthesized for the targeted delivery of GEM to integrin αvß3, overexpressing cancer cells to increase the stability as well as the tumor delivery of GEM and minimize common side effects associated with GEM treatment. Competitive cell uptake experiments demonstrated that conjugate TC113 could be internalized by A549 cells through integrin αvß3. Among the synthesized conjugates, TC113 bearing the carbamate linker was stable in human plasma and was further assessed in an in vivo pharmacokinetic study. TC113 appeared to be relatively stable, releasing GEM slowly into blood, while it showed potent antiproliferative properties against WM266.4 and A549 cells. The encouraging data presented in this study with respect to TC113 provide a promising keystone for further investigation of this GEM conjugate with potential future clinical applications.


Subject(s)
Deoxycytidine/analogs & derivatives , Integrins/chemistry , Lung Neoplasms/drug therapy , Peptides, Cyclic/chemistry , A549 Cells , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Cell Proliferation , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Gemcitabine
3.
Cancers (Basel) ; 13(8)2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33920884

ABSTRACT

Pioneering studies on tumor and immune cell interactions have highlighted immune checkpoint inhibitors (ICIs) as revolutionizing interventions for the management of NSCLC, typically combined with traditional MTD chemotherapies, which usually lead to toxicities and resistance to treatment. Alternatively, MTR chemotherapy is based on the daily low dose administration of chemotherapeutics, preventing tumor growth indirectly by targeting the tumor microenvironment. The effects of MTR administration of an oral prodrug of gemcitabine (OralGem), alone or with anti-PD1, were evaluated. Relevant in vitro and in vivo models were developed to investigate the efficacy of MTR alone or with immunotherapy and the potential toxicities associated with each dosing scheme. MTR OralGem restricted tumor angiogenesis by regulating thrombospondin-1 (TSP-1) and vascular endothelial growth factor A (VEGFA) expression. MTR OralGem enhanced antitumor immunity by increasing T effector responses and cytokine release, concomitant with dampening regulatory T cell populations. Promising pharmacokinetic properties afforded minimized blood and thymus toxicity and favorable bioavailability upon MTR administration compared to MTD. The combination of MTR OralGem with immunotherapy was shown to be highly efficacious and tolerable, illuminating it as a strong candidate therapeutic scheme for the treatment of NSCLC.

4.
Eur J Med Chem ; 211: 113018, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33223264

ABSTRACT

Peptide-drug conjugates (PDCs) are gaining considerable attention as anti-neoplastic agents. However, their development is often laborious and time-consuming. Herein, we have developed and preclinically evaluated three PDCs with gemcitabine as the anticancer cytotoxic unit and D-Lys6-GnRH (gonadotropin-releasing hormone; GnRH) as the cancer-targeting unit. These units were tethered via acid-labile programmable linkers to guide a differential drug release rate from the PDC through a combination of ester or amide and "click" type oxime ligations. The pro-drugs were designed to enable the selective targeting of malignant tumor cells with linker guided differential drug release rates. We exploited the oxime bond responsiveness against the acidic pH of the tumor microenvironment and the GnRH endocytosis via the GnRH-R GPCR which is overexpressed on cancer cells. The challenging metabolic properties of gemcitabine were addressed during design of the PDCs. We developed a rapid (1 hour) and cost-effective "click" oxime bond ligation platform to assemble in one-pot the 3 desired PDCs that does not require purification, surpassing traditional time-ineffective and low yield methods. The internalization of the tumor-homing peptide unit in cancer cells, overexpressing the GnRH-R, was first validated through confocal laser microscopy and flow cytometry analysis. Subsequently, the three PDCs were evaluated for their in vitro antiproliferative effect in prostate cancer cells. Their stability and the release of gemcitabine over time were monitored in vitro in cell culture and in human plasma using LC-MS/MS. We then assessed the ability of the developed PDCs to internalize in prostate cancer cells and to release gemcitabine. The most potent analog, designated GOXG1, was used for pharmacokinetic studies in mice. The metabolism of GOXG1 was examined in liver microsomes, as well as in buffers mimicking the pH of intracellular organelles, resulting in the identification of two metabolites. The major metabolite at low pH emanated from the cleavage of the pH-labile oxime bond, validating our design approach. NMR spectroscopy and in vitro radioligand binding assays were exploited for GOXG1 to validate that upon conjugating the drug to the peptide, the peptide microenvironment responsible for its GnRH-R binding is not perturbed and to confirm its high binding potency to the GnRH-R. Finally, the binding of GOXG1 to the GnRH-R and the associated elicitation of testosterone release in mice were also determined. The facile platform established herein for the rapid assembly of PDCs with linker controllable characteristics from aldehyde and aminooxy units through rapid "click" oxime ligation, that does not require purification steps, could pave the way for a new generation of potent cancer therapeutics, diagnostics and theranostics.


Subject(s)
Deoxycytidine/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Oximes/pharmacology , Prodrugs/pharmacology , Prostatic Neoplasms/drug therapy , Receptors, LHRH/agonists , Animals , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Development , Gonadotropin-Releasing Hormone/administration & dosage , Gonadotropin-Releasing Hormone/chemistry , Humans , Hydrogen-Ion Concentration , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Oximes/administration & dosage , Oximes/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured , Gemcitabine
5.
Eur J Med Chem ; 176: 393-409, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31125894

ABSTRACT

Novel substituted purine isosters, were designed and synthesized as potential inhibitors of the Epidermal Growth Factor Receptor (EGFR). The compounds were rationally designed through bioisosteric replacement of the central quinazoline core of lapatinib, an approved drug that inhibits both EGFR and HER2, another important member of this family of receptors. The new target molecules were evaluated as inhibitors of receptor phosphorylation at the cellular level, for their direct inhibitory action on the intracellular receptor kinase domain and for their cytotoxicity against the non-small cell lung cancer cell line A549 and breast cancer HCC1954, cell lines which are associated with overexpression of EGFR and HER2, respectively. The most potent derivatives were further studied for their cellular uptake levels and in vivo pharmacokinetic properties. One compound (23) displayed a noteworthy pharmacokinetic profile, and higher intracellular accumulation in comparison to lapatinib in the A549 cells, possibly due to its higher lipophilicity. This lead compound (23) was assessed for its efficacy in an EGFR positive xenograft model, where it successfully inhibited tumor growth, with a similar efficacy with that of lapatinib and with minimal phenotypic toxicity.


Subject(s)
Antineoplastic Agents/therapeutic use , Lapatinib/analogs & derivatives , Lapatinib/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Purines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Catalytic Domain , Cell Line, Tumor , Female , Humans , Lapatinib/chemical synthesis , Lapatinib/pharmacokinetics , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Purines/chemical synthesis , Purines/chemistry , Purines/pharmacokinetics , Receptor, ErbB-2/chemistry
6.
Eur J Med Chem ; 166: 256-266, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30716713

ABSTRACT

Peptide-drug conjugates have emerged as a potent approach to enhance the targeting and pharmacokinetic profiles of drugs. However, the impact of the linker unit has not been explored/exploited in depth. Gemcitabine (dFdC) is an anticancer agent used against a variety of solid tumours. Despite its potency, gemcitabine suffers mostly due to its unspecific toxicity, lack of targeting and rapid metabolic inactivation. To minimize these limitations and enable its targeting to tumours overexpressing the GnRH receptor, we examined the peptide-drug conjugation approach. Our design hypothesis was driven by the impact that the linker unit could have on the peptide-drug conjugate efficacy. Along these lines, in order to exploit the potential to manipulate the potency of gemcitabine through altering the linker unit we constructed three different novel peptide-drug conjugates assembled of gemcitabine, the tumour-homing peptide D-Lys6-GnRH and modified linker building blocks. Specifically, the linker was sculpted to either allow slow drug release (utilizing carbamate bond) or rapid disassociation (using amide and ester bonds). Notably, the new analogues possessed up to 95.5-fold enhanced binding affinity for the GnRH receptor (GnRH-R) compared to the natural peptide ligand D-Lys6-GnRH. Additionally, their in vitro cytotoxicity was evaluated in four different cancer cell lines. Their cellular uptake, release of gemcitabine and inactivation of gemcitabine to its inactive metabolite (dFdU) was explored in a representative cell line. In vitro stability and the consequent drug release were evaluated in cell culture medium and human plasma. In vivo pharmacokinetic studies were performed in mice, summarizing the relative stability of the three conjugates and the released levels of gemcitabine in comparison with dFdU. These studies suggest that the fine tuning of the linkage within a peptide-drug conjugate affects the drug release rate and its overall pharmaceutical profile. This could eventually emerge as an intriguing medicinal chemistry approach to optimize bio-profiles of prodrugs.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Liberation , Gonadotropin-Releasing Hormone/chemistry , Lysine/chemistry , Prodrugs/metabolism , Animals , Cell Proliferation/drug effects , Deoxycytidine/chemistry , Deoxycytidine/metabolism , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Drug Stability , Humans , Intracellular Space/metabolism , MCF-7 Cells , Mice , Receptors, LHRH/metabolism , Gemcitabine
7.
Mater Sci Eng C Mater Biol Appl ; 79: 605-612, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28629059

ABSTRACT

Nanostructured delivery and diagnostic systems that induces specific targeting properties by exploiting the local physicochemical tumour characteristics will be evaluated is the present work. It is well known that cancer cells have specific physicochemical characteristics, which can be taken into consideration for the design of a broad spectrum of drug delivery systems (DDS). Some of those characteristics including the different temperature environment their susceptibility when temperature ranges between 40 and 43°C where cell apoptosis is induced, the intra- and extra-cellular pH which varies from 6.0 to 6.8, for cancer cells, and 6.5 to 7.4 for normal cells respectively, (lysosomes acidic pH ranges 4-5). Additional significant factors are the overexpressed receptors on the tumour surface. Loading and release studies were carried out by using the anthracycline drug Doxorubicin and their cytotoxicity was evaluated by using the MTT assay in healthy and diseased cell lines. The highlight of this work is the in vitro and in vivo studies which were performed in order to evaluate different nanostructures as for their biodistribution, pharmacokinetic and toxicity per se.


Subject(s)
Nanostructures , Antibiotics, Antineoplastic , Doxorubicin , Drug Delivery Systems , Humans , Tissue Distribution
8.
Oncotarget ; 8(23): 37250-37262, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28422745

ABSTRACT

The clinical efficacy of antiangiogenic small molecules (e.g., sunitinib) in breast carcinoma has largely failed with substantial off-target toxicity. We rationally designed and evaluated preclinically a novel sunitinib analogue, SAP, with favourable pharmacological properties and the ability to be readily conjugated to a targeting peptide or antibody for active tumour targeting.SAP was evaluated in silico and in vitro in order to verify target engagement (e.g., VEGFR2). Pharmacokinetic and biodistribution parameters were determined in mice using LC-MS/MS. SAP efficacy was tested in two breast cancer xenograft and two syngeneic animal models and pharmacodynamic evaluation was accomplished using phosphokinase assays and immunohistochemistry. Cardiac and blood toxicity of SAP were also monitored.SAP retained the antiangiogenic and cytotoxic properties of the parental molecule with an increased blood exposure and tumor accumulation compared to sunitinib. SAP proved efficacious in all animal models. Tumors from SAP treated animals had significantly decreased Ki-67 and CD31 markers and reduced levels of phosphorylated AKT, ERK and S6 compared to vehicle treated animals. In mice dosed with SAP there was negligible hematotoxicity, while cardiac function measurements showed a reduction in the percentage left ventricular fractional shortening compared to vehicle treated animals.In conclusion, SAP is a novel rationally designed conjugatable small antiangiogenic molecule, efficacious in preclinical models of breast cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Indoles/therapeutic use , Neoplasms, Experimental/drug therapy , Angiogenesis Inhibitors/chemical synthesis , Animals , Cell Line, Tumor , Cell Proliferation , Female , Human Umbilical Vein Endothelial Cells , Humans , Indoles/chemical synthesis , Indoles/chemistry , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasms, Experimental/pathology , Oxindoles , Pyrroles/chemistry , Pyrroles/therapeutic use , Sunitinib , Tumor Burden , Tumor Microenvironment , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 114(15): 3999-4004, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28348207

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic (DAergic) neurons in the substantia nigra and the gradual depletion of dopamine (DA). Current treatments replenish the DA deficit and improve symptoms but induce dyskinesias over time, and neuroprotective therapies are nonexistent. Here we report that Nuclear receptor-related 1 (Nurr1):Retinoid X receptor α (RXRα) activation has a double therapeutic potential for PD, offering both neuroprotective and symptomatic improvement. We designed BRF110, a unique in vivo active Nurr1:RXRα-selective lead molecule, which prevents DAergic neuron demise and striatal DAergic denervation in vivo against PD-causing toxins in a Nurr1-dependent manner. BRF110 also protects against PD-related genetic mutations in patient induced pluripotent stem cell (iPSC)-derived DAergic neurons and a genetic mouse PD model. Remarkably, besides neuroprotection, BRF110 up-regulates tyrosine hydroxylase (TH), aromatic l-amino acid decarboxylase (AADC), and GTP cyclohydrolase I (GCH1) transcription; increases striatal DA in vivo; and has symptomatic efficacy in two postneurodegeneration PD models, without inducing dyskinesias on chronic daily treatment. The combined neuroprotective and symptomatic effects of BRF110 identify Nurr1:RXRα activation as a potential monotherapeutic approach for PD.


Subject(s)
Antiparkinson Agents/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Parkinson Disease/drug therapy , Retinoid X Receptor alpha/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antiparkinson Agents/chemistry , Antiparkinson Agents/pharmacokinetics , Brain/drug effects , Cell Line , Disease Models, Animal , Dopamine/genetics , Drug Stability , Humans , Male , Mice, Inbred BALB C , Molecular Targeted Therapy , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Multimerization , Rats , Retinoid X Receptor alpha/agonists , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/genetics
10.
Mol Pharm ; 14(3): 674-685, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28099809

ABSTRACT

Gemcitabine is a clinically established anticancer agent potent in various solid tumors but limited by its rapid metabolic inactivation and off-target toxicity. We have previously generated a metabolically superior to gemcitabine molecule (GSG) by conjugating gemcitabine to a gonadotropin releasing hormone receptor (GnRH-R) ligand peptide and showed that GSG was efficacious in a castration resistant prostate cancer (CRPC) animal model. The current article provides an in-depth metabolic and mechanistic study of GSG, coupled with toxicity assays that strengthen the potential role of GSG in the clinic. LC-MS/MS based approaches were employed to delineate the metabolism of GSG, its mechanistic cellular uptake, and release of gemcitabine and to quantitate the intracellular levels of gemcitabine and its metabolites (active dFdCTP and inactive dFdU) resulting from GSG. The GnRH-R agonistic potential of GSG was investigated by quantifying the testosterone levels in animals dosed daily with GSG, while an in vitro colony forming assay together with in vivo whole blood measurements were performed to elucidate the hematotoxicity profile of GSG. Stability showed that the major metabolite of GSG is a more stable nonapeptide that could prolong gemcitabine's bioavailability. GSG acted as a prodrug and offered a metabolic advantage compared to gemcitabine by generating higher and steadier levels of dFdCTP/dFdU ratio, while intracellular release of gemcitabine from GSG in DU145 CRPC cells depended on nucleoside transporters. Daily administrations in mice showed that GSG is a potent GnRH-R agonist that can also cause testosterone ablation without any observed hematotoxicity. In summary, GSG could offer a powerful and unique pharmacological approach to prostate cancer treatment: a single nontoxic molecule that can be used to reach the tumor site selectively with superior to gemcitabine metabolism, biodistribution, and safety while also agonistically ablating testosterone levels.


Subject(s)
Deoxycytidine/analogs & derivatives , Peptides/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Deoxycytidine/pharmacology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Prodrugs/pharmacology , Prostatic Neoplasms, Castration-Resistant/metabolism , Rats, Wistar , Receptors, LHRH/metabolism , Tissue Distribution/physiology , Tumor Cells, Cultured , Gemcitabine
11.
Cardiovasc Intervent Radiol ; 39(10): 1379-91, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27393274

ABSTRACT

PURPOSE: This study examines safety, efficacy, and pharmacokinetics of chemoembolization with loadable microspheres ≤100 µm for hepatocellular carcinoma. MATERIALS AND METHODS: A pilot safety study was performed in 19 patients with size and dose escalation and then 52 patients were enrolled prospectively and randomly assigned to chemoembolization with TANDEM™ loaded with 150 or 100 mg of doxorubicin. RESULTS: The mean diameter of the tumors was 7.28 ± 2.09 cm (range 4-12) and distribution dominant/multiple 51.9/48.1 %. Child A/B distribution was 32/20 (61.5/38.5 %) and etiology HBV/HCV/HBV/HCV-hemochromatosis was 61.6/9.6/9.6/15.4 %. Twenty-five patients were assigned in the low and 27 in the high loading group. There was 1.92 % thirty-day mortality due to lesion rupture. Biliary damage was seen in 3 patients (5.7 %) in the high loading. Mean maximum plasma concentration of doxorubicin C max ± SD was 284.9 ± 276.2 ng/mL for the high and 108.5 ± 77.6 ng/mL for the low loading (p < 0.001). According to m-RECIST overall objective response after two sessions reached 61.22 and 63.82 % at 6 months. Notably, complete target lesion response (CR) after the second session was observed in 28.57 % and maintained in 23.40 % at 6 months. No statistical differences in the local response rates were observed between the two loading groups. Overall survival (OS) at 6 months, 1 , 2, and 3 years was 98.08, 92.3, 88.46, and 82.6 %, respectively. OS and Progression-Free Survival did not demonstrate statistical significance between the two loading groups. CONCLUSION: Initial evidence shows that (a) TANDEM™ achieves high rates of local response and mid-term survival, (b) high loading provides no clinical benefit and is associated with biliary toxicity.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/methods , Doxorubicin/pharmacokinetics , Liver Neoplasms/therapy , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/administration & dosage , Chemoembolization, Therapeutic/adverse effects , Disease-Free Survival , Doxorubicin/administration & dosage , Female , Humans , Male , Microspheres , Middle Aged , Pilot Projects , Treatment Outcome
13.
Cancer Res ; 76(5): 1181-92, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26645560

ABSTRACT

The potential to heighten the efficacy of antiangiogenic agents was explored in this study based on active targeting of tumor cells overexpressing the gonadotropin-releasing hormone receptor (GnRH-R). The rational design pursued focused on five analogues of a clinically established antiangiogenic compound (sunitinib), from which a lead candidate (SAN1) was conjugated to the targeting peptide [d-Lys(6)]-GnRH, generating SAN1GSC. Conjugation of SAN1 did not disrupt any of its antiangiogenic or cytotoxic properties in GnRH-R-expressing prostate and breast tumor cells. Daily SAN1GSC treatments in mouse xenograft models of castration-resistant prostate cancer resulted in significant tumor growth delay compared with equimolar SAN1 or sunitinib alone. This efficacy correlated with inhibited phosphorylation of AKT and S6, together with reduced Ki-67 and CD31 expression. The superior efficacy of the peptide-drug conjugate was also attributed to the finding that higher amounts of SAN1 were delivered to the tumor site (∼4-fold) following dosing of SAN1GSC compared with equimolar amounts of nonconjugated SAN1. Importantly, treatment with SAN1GSC was associated with minimal hematotoxicity and cardiotoxicity based on measurements of the left ventricular systolic function in treated mice. Our results offer preclinical proof-of-concept for SAN1GSC as a novel molecule that selectively reaches the tumor site and downregulates angiogenesis with negligible cardiotoxicity, thus encouraging its further clinical development and evaluation.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Gonadotropin-Releasing Hormone/pharmacology , Indoles/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Pyrroles/pharmacology , Animals , Cell Proliferation/drug effects , Drug Design , Humans , Male , Mice , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, LHRH/analysis , Sunitinib , Xenograft Model Antitumor Assays
14.
Bioconjug Chem ; 25(4): 813-23, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24661240

ABSTRACT

Gemcitabine, a drug with established efficacy against a number of solid tumors, has therapeutic limitations due to its rapid metabolic inactivation. The aim of this study was the development of an innovative strategy to produce a metabolically stable analogue of gemcitabine that could also be selectively delivered to prostate cancer (CaP) cells based on cell surface expression of the Gonadotropin Releasing Hormone-Receptor (GnRH-R). The synthesis and evaluation of conjugated molecules, consisting of gemcitabine linked to a GnRH agonist, is presented along with results in androgen-independent prostate cancer models. NMR and ligand binding assays were employed to verify conservation of microenvironments responsible for binding of novel GnRH-gemcitabine conjugates to the GnRH-R. In vitro cytotoxicity, cellular uptake, and metabolite formation of the conjugates were examined in CaP cell lines. Selected conjugates were efficacious in the in vitro assays with one of them, namely, GSG, displaying high antiproliferative activity in CaP cell lines along with significant metabolic and pharmacokinetic advantages in comparison to gemcitabine. Finally, treatment of GnRH-R positive xenografted mice with GSG showed a significant advantage in tumor growth inhibition when compared to gemcitabine.


Subject(s)
Deoxycytidine/analogs & derivatives , Drug Delivery Systems , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Cell Proliferation/drug effects , Deoxycytidine/chemistry , Deoxycytidine/metabolism , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Gonadotropin-Releasing Hormone/pharmacokinetics , Gonadotropin-Releasing Hormone/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Molecular Structure , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, LHRH/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
15.
Cardiovasc Intervent Radiol ; 37(1): 165-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24263774

ABSTRACT

BACKGROUND: This study examined the safety, pharmacokinetics, and efficacy of transarterial chemoembolization of hepatocellular carcinoma (HCC) using a newly developed size of a superabsorbent polymer drug-eluting embolic material. METHODS: Forty-five patients with documented HCC (Child-Pugh score A/B: 55.5 %/44.5 %) were embolized with HepaSphere microspheres 30-60 µm with escalation of lesion, dose, and frequency of re-embolization. Local response was evaluated with modified response evaluation criteria in solid tumors (mRECIST). Plasma levels of doxorubicin were measured in 24 patients at baseline and at 5, 20, 40, 60, and 120 min, at 6, 24, and 48 h, and at 7 days, respectively, to determine doxorubicin in plasma (Cmax) and area under the curve (AUC). Measurements of three patients who underwent lipiodol-based conventional chemoembolization (c-TACE) were also performed. RESULTS: TACE with HepaSphere was well tolerated with an acceptable safety profile and no 30-day mortality. Response rates were calculated on intention-to-treat basis with complete response (CR) in 17.8 % reaching 22.2 % for the target lesion. Overall partial response (PR) was seen in 51.1 %, stable disease in 20 %, and progressive disease in 11.1 % of patients. Overall objective response (CR + PR), including patients treated at all dosages of doxorubicin, was seen in 68.9 % of cases. After a median follow-up of 15.6 months, 1-year survival is 100 %. Doxorubicin AUC was significantly lower in patients with HepaSphere 30-60 µm (35,195 ± 27,873 ng × min/ml) than in patients with conventional TACE (103,960 ± 16,652 ng × min/ml; p = 0.009). Cmax was also significantly lower with HepaSphere 30-60 µm (83.9 ± 32.1 ng/ml) compared with c-TACE (761.3 ± 58.8 ng/ml; p = 0.002). CONCLUSION: HepaSphere 30-60 µm is an effective drug-eluting embolic material with a favourable pharmacokinetic profile.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/methods , Doxorubicin/administration & dosage , Ethiodized Oil/administration & dosage , Liver Neoplasms/therapy , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacokinetics , Carcinoma, Hepatocellular/diagnosis , Contrast Media , Diagnostic Imaging , Doxorubicin/pharmacokinetics , Drug Carriers , Ethiodized Oil/pharmacokinetics , Female , Humans , Liver Function Tests , Liver Neoplasms/diagnosis , Male , Microspheres , Middle Aged , Phospholipids , Polymers , Prospective Studies , Sulfur Hexafluoride , Survival Rate , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...