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










Publication year range
1.
ACS Pharmacol Transl Sci ; 4(2): 953-965, 2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33860213

ABSTRACT

Lipophilicity is explored in the biodistribution (BD), pharmacokinetics (PK), radiation dosimetry (RD), and toxicity of an internally administered targeted alpha-particle therapy (TAT) under development for the treatment of metastatic melanoma. The TAT conjugate is comprised of the chelator DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetate), conjugated to melanocortin receptor 1 specific peptidic ligand (MC1RL) using a linker moiety and chelation of the 225Ac radiometal. A set of conjugates were prepared with a range of lipophilicities (log D 7.4 values) by varying the chemical properties of the linker. Reported are the observations that higher log D 7.4 values are associated with decreased kidney uptake, decreased absorbed radiation dose, and decreased kidney toxicity of the TAT, and the inverse is observed for lower log D 7.4 values. Animals administered TATs with lower lipophilicities exhibited acute nephropathy and death, whereas animals administered the highest activity TATs with higher lipophilicities lived for the duration of the 7 month study and exhibited chronic progressive nephropathy. Changes in TAT lipophilicity were not associated with changes in liver uptake, dose, or toxicity. Significant observations include that lipophilicity correlates with kidney BD, the kidney-to-liver BD ratio, and weight loss and that blood urea nitrogen (BUN) levels correlated with kidney uptake. Furthermore, BUN was identified as having higher sensitivity and specificity of detection of kidney pathology, and the liver enzyme alkaline phosphatase (ALKP) had high sensitivity and specificity for detection of liver damage associated with the TAT. These findings suggest that tuning radiopharmaceutical lipophilicity can effectively modulate the level of kidney uptake to reduce morbidity and improve both safety and efficacy.

2.
Eur J Nucl Med Mol Imaging ; 48(11): 3408-3421, 2021 10.
Article in English | MEDLINE | ID: mdl-33772332

ABSTRACT

PURPOSE: There is significant interest in the development of targeted alpha-particle therapies (TATs) for treatment of solid tumors. The metal chelator-peptide conjugate, DOTA-TATE, loaded with the ß-particle emitting radionuclide 177Lu ([177Lu]Lu-DOTA-TATE) is now standard care for neuroendocrine tumors that express the somatostatin receptor 2 (SSTR2) target. A recent clinical study demonstrated efficacy of the corresponding [225Ac]Ac-DOTA-TATE in patients that were refractory to [177Lu]Lu-DOTA-TATE. Herein, we report the radiosynthesis, toxicity, biodistribution (BD), radiation dosimetry (RD), and efficacy of [225Ac]Ac-DOTA-TATE in small animal models of lung neuroendocrine neoplasms (NENs). METHODS: [225Ac]Ac-DOTA-TATE was synthesized and characterized for radiochemical yield, purity and stability. Non-tumor-bearing BALB/c mice were tested for toxicity and BD. Efficacy was determined by single intravenous injection of [225Ac]Ac-DOTA-TATE into SCID mice-bearing human SSTR2 positive H727 and H69 lung NENs. RD was calculated using the BD data. RESULTS: [225Ac]Ac-DOTA-TATE was synthesized with 98% yield, 99.8% purity, and displayed 97% stability after 2 days incubation in human serum at 37 °C. All animals in the toxicity study appeared healthy 5 months post injection with no indications of toxicity, except that animals that received ≥111 kBq of [225Ac]Ac-DOTA-TATE had chronic progressive nephropathy. BD studies revealed that the primary route of elimination is by the renal route. RD calculations determined pharmacokinetics parameters and absorbed α-emission dosages from 225Ac and its daughters. For both tumor models, a significant tumor growth delay and time to experimental endpoint were observed following a single administration of [225Ac]Ac-DOTA-TATE relative to controls. CONCLUSIONS: These results suggest significant potential for the clinical translation of [225Ac]Ac-DOTA-TATE for lung NENs.


Subject(s)
Lung Neoplasms , Organometallic Compounds , Animals , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, SCID , Octreotide/therapeutic use , Octreotide/toxicity , Organometallic Compounds/therapeutic use , Organometallic Compounds/toxicity , Radiopharmaceuticals/therapeutic use , Radiopharmaceuticals/toxicity , Tissue Distribution
3.
Mol Pharm ; 17(11): 4180-4188, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32960613

ABSTRACT

Targeted α particle therapy (TAT) is ideal for treating disease while minimizing damage to surrounding nontargeted tissues due to short path length and high linear energy transfer (LET). We developed a TAT for metastatic uveal melanoma, targeting the melanocortin-1 receptor (MC1R), which is expressed in 94% of uveal melanomas. Two versions of the therapy are being investigated: 225Ac-DOTA-Ahx-MC1RL (225Ac-Ahx) and 225Ac-DOTA-di-d-Glu-MC1RL (225Ac-di-d-Glu). The biodistribution (BD) from each was studied and a multicompartment pharmacokinetic (PK) model was developed to describe drug distribution rates. Two groups of 16 severe combined immunodeficient (SCID) mice bearing high MC1R expressing tumors were intravenously injected with 225Ac-Ahx or 225Ac-di-d-Glu. After injection, four groups (n = 4) were euthanized at 24, 96, 144, and 288 h time points for each cohort. Tumors and 13 other organs were harvested at each time point. Isomeric γ spectra were measured in tissue samples using a scintillation γ detector and converted to α activity using factors for γ ray abundance per α decay. Time activity curves were calculated for each organ. A five-compartment PK model was built with the following compartments: blood, tumor, normal tissue, kidney, and liver. This model is characterized by a system of five ordinary differential equations using mass action kinetics, which describe uptake, intercompartmental transitions, and clearance rates. The ordinary differential equations were simultaneously solved and fit to experimental data using a genetic algorithm for optimization. The BD data show that both compounds have minimal distribution to organs at risk other than the kidney and liver. The PK parameter estimates had less than 5% error. From these data, 225Ac-Ahx showed larger and faster uptake in the liver. Both compounds had comparable uptake and clearance rates for other compartments. The BD and PK behavior for two targeted radiopharmaceuticals were investigated. The PK model fit the experimental data and provided insight into the kinetics of the compounds systematically.


Subject(s)
Alpha Particles/therapeutic use , Melanoma, Experimental/drug therapy , Melanoma/drug therapy , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Uveal Neoplasms/drug therapy , alpha-MSH/administration & dosage , alpha-MSH/pharmacokinetics , Animals , Cell Line, Tumor , Ligands , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, SCID , Molecular Targeted Therapy/methods , Receptor, Melanocortin, Type 1/metabolism , Tissue Distribution , Treatment Outcome , Tumor Burden/drug effects , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
World J Clin Oncol ; 11(4): 169-179, 2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32355639

ABSTRACT

Triple-negative breast cancer (TNBC) is defined as a type of breast cancer with lack of expression of estrogen receptor, progesterone receptor and human epidermal growth factor 2 protein. In comparison to other types of breast cancer, TNBC characterizes for its aggressive behavior, more prone to early recurrence and a disease with poor response to molecular target therapy. Although TNBC is identified in only 25%-30% of American breast cancer cases annually, these tumors continue to be a therapeutic challenge for clinicians for several reasons: Tumor heterogeneity, limited and toxic systemic therapy options, and often resistance to current standard therapy, characterized by progressive disease on treatment, residual tumor after cytotoxic chemotherapy, and early recurrence after complete surgical excision. Cell-surface targeted therapies have been successful for breast cancer in general, however there are currently no approved cell-surface targeted therapies specifically indicated for TNBC. Recently, several cell-surface targets have been identified as candidates for treatment of TNBC and associated targeted therapies are in development. The purpose of this work is to review the current clinical challenges posed by TNBC, the therapeutic approaches currently in use, and provide an overview of developing cell surface targeting approaches to improve outcomes for treatment resistant TNBC.

5.
Molecules ; 24(23)2019 Nov 26.
Article in English | MEDLINE | ID: mdl-31779154

ABSTRACT

Targeted alpha-particle therapy (TAT) aims to selectively deliver radionuclides emitting α-particles (cytotoxic payload) to tumors by chelation to monoclonal antibodies, peptides or small molecules that recognize tumor-associated antigens or cell-surface receptors. Because of the high linear energy transfer (LET) and short range of alpha (α) particles in tissue, cancer cells can be significantly damaged while causing minimal toxicity to surrounding healthy cells. Recent clinical studies have demonstrated the remarkable efficacy of TAT in the treatment of metastatic, castration-resistant prostate cancer. In this comprehensive review, we discuss the current consensus regarding the properties of the α-particle-emitting radionuclides that are potentially relevant for use in the clinic; the TAT-mediated mechanisms responsible for cell death; the different classes of targeting moieties and radiometal chelators available for TAT development; current approaches to calculating radiation dosimetry for TATs; and lead optimization via medicinal chemistry to improve the TAT radiopharmaceutical properties. We have also summarized the use of TATs in pre-clinical and clinical studies to date.


Subject(s)
Alpha Particles/therapeutic use , Neoplasms/radiotherapy , Radiopharmaceuticals/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Humans , Radioisotopes/therapeutic use , Radiometry/methods
6.
J Nucl Med ; 60(8): 1124-1133, 2019 08.
Article in English | MEDLINE | ID: mdl-30733316

ABSTRACT

New effective therapies are greatly needed for metastatic uveal melanoma, which has a very poor prognosis with a median survival of less than 1 y. The melanocortin 1 receptor (MC1R) is expressed in 94% of uveal melanoma metastases, and a MC1R-specific ligand (MC1RL) with high affinity and selectivity for MC1R was previously developed. Methods: The 225Ac-DOTA-MC1RL conjugate was synthesized in high radiochemical yield and purity and was tested in vitro for biostability and for MC1R-specific cytotoxicity in uveal melanoma cells, and the lanthanum-DOTA-MC1RL analog was tested for binding affinity. Non-tumor-bearing BALB/c mice were tested for maximum tolerated dose and biodistribution. Severe combined immunodeficient mice bearing uveal melanoma tumors or engineered MC1R-positive and -negative tumors were studied for biodistribution and efficacy. Radiation dosimetry was calculated using mouse biodistribution data and blood clearance kinetics from Sprague-Dawley rat data. Results: High biostability, MC1R-specific cytotoxicity, and high binding affinity were observed. Limiting toxicities were not observed at even the highest administered activities. Pharmacokinetics and biodistribution studies revealed rapid blood clearance (<15 min), renal and hepatobillary excretion, MC1R-specific tumor uptake, and minimal retention in other normal tissues. Radiation dosimetry calculations determined pharmacokinetics parameters and absorbed α-emission dosages from 225Ac and its daughters. Efficacy studies demonstrated significantly prolonged survival and decreased metastasis burden after a single administration of 225Ac-DOTA-MC1RL in treated mice relative to controls. Conclusion: These results suggest significant potential for the clinical translation of 225Ac-DOTA-MC1RL as a novel therapy for metastatic uveal melanoma.


Subject(s)
Melanoma/radiotherapy , Molecular Targeted Therapy , Receptor, Melanocortin, Type 1/chemistry , Uveal Neoplasms/radiotherapy , Alpha Particles , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chelating Agents/chemistry , Female , Humans , Lanthanoid Series Elements/chemistry , Male , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Metastasis , Neoplasm Transplantation , Prognosis , Radiometry , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley
7.
Mol Imaging Biol ; 18(2): 219-31, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26276155

ABSTRACT

PURPOSE: Hypoxia is commonly observed in regions of primary tumors and metastases, and is associated with resistance to treatment, more aggressive tumor phenotypes and poor prognosis. Reliable and validated imaging biomarkers of hypoxia are needed for pre-clinical studies and clinical use. Expression of cell-surface carbonic anhydrases IX and XII (CAIX and CAXII) in tumor cells has been associated with tumor hypoxia. CAIX and CAXII specific antibodies conjugated to fluorescent dye were evaluated for the non-invasive detection of hypoxia in vivo. PROCEDURES: Human breast cancer cell lines (MCF10A, DCIS, MCF7, ZR-75.1 and MDA-mb231) were characterized for CAIX and CAXII expression by real-time RT-PCR and immunocytochemistry (ICC) under normoxic and hypoxic conditions. Immunohistochemical (IHC) staining of CAIX, CAXII and the commercially available exogenous hypoxia marker, pimonidazole, was performed using sections of ZR-75.1 and MDA-mb-231 orthotopic breast cancer xenograft tumors from nude mice. In vivo fluorescence imaging of ZR-75.1 tumors in animals housed at varied levels of oxygen was used to quantify the relative uptake of the CAIX and CAXII agents and a commercially available sulfonamide-based agent. Corresponding tumor sections were IHC stained for CAIX, CAXII and pimonidazole. RESULTS: CAIX mRNA expression was significantly higher (p < 0.05) in hypoxia for all cell lines, which was in agreement with protein expression by ICC. CAXII expression was mixed, with a modest hypoxia-related increase in two cell lines (p < 0.05) and no change in others. Quantified IHC staining of ZR-75.1 and MDA-mb-231 tumor sections showed that CAIX and CAXII expression was elevated in regions with pimonidazole staining, but CAXII levels were lower than CAIX. Tumor uptake of the CAIX targeted agent, and IHC staining of CAIX and pimonidazole in corresponding tumor sections were correlated, and co-registered, and shown to be significantly elevated by level of oxygenation (p < 0.001): hypoxia > normoxia > hyperoxia. However, the CAXII and sulfonamide agents were not significantly correlated with hypoxia. CONCLUSION: These studies suggest that the fluorescently labeled CAIX-specific agent is a more robust indicator of hypoxia in vivo compared to the CAXII-specific agent or the agent specific to the CA active site.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrases/metabolism , Molecular Imaging/methods , Oxygen/pharmacology , Tumor Hypoxia/drug effects , Tumor Hypoxia/genetics , Analysis of Variance , Animals , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carbonic Anhydrase IX/genetics , Carbonic Anhydrases/genetics , Cell Line, Tumor , Female , Fluorescence , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tomography , Xenograft Model Antitumor Assays
8.
Nat Commun ; 6: 8752, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26658462

ABSTRACT

Early cancers are avascular and hence, profoundly acidic. Pre-malignant cells must adapt to acidosis to thrive in this hostile microenvironment. Here, we investigate MCF-7 cells that are adapted to grow in acidic conditions using SILAC proteomics and we reveal a significant upregulation of lysosomal proteins. Prominent among these is LAMP2 that functions to protect lysosomal membranes from acid proteolysis. LAMP2 upregulation by acidosis is confirmed both in vitro and in vivo. Furthermore, we show that the depletion of LAMP2 is sufficient to increase acidosis-mediated toxicity. In breast cancer patient samples, there is a high correlation of LAMP2 mRNA and protein expression with progression. We also observe that LAMP2 is located at the plasma membrane in clinical samples and this redistribution is acid-induced in vitro. Our findings suggest a potential adaptive mechanism, wherein cells chronically exposed to an acidic environment translocate lysosomal proteins to their surface, thus protecting the plasmalemma from acid-induced hydrolysis.


Subject(s)
Cell Membrane/metabolism , Gene Expression Regulation, Neoplastic/physiology , Lysosomal-Associated Membrane Protein 2/metabolism , Neoplasms/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Lysosomal-Associated Membrane Protein 2/genetics , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Protein Array Analysis , Proteomics
9.
Front Immunol ; 5: 429, 2014.
Article in English | MEDLINE | ID: mdl-25368611

ABSTRACT

Toll-like receptors (TLRs) are expressed by immune cells, intestinal epithelium, and tumor cells. In the homeostatic setting, they help to regulate control over invading pathogens and maintain the epithelial lining of the large and small intestines. Aberrant expression of certain TLRs by tumor cells can induce growth inhibition while others contribute to tumorigenesis and progression. Activation of these TLRs can induce inflammation, tumor cell proliferation, immune evasion, local invasion, and distant metastasis. These TLR-influenced behaviors have similarities with properties observed in leukocytes, suggesting that tumors may be hijacking immune programs to become more aggressive. The concept of epithelial to leucocytic-transition (ELT) is proposed, akin to epithelial to mesenchymal transition, in which tumors develop the ability to activate leucocytic traits otherwise inaccessible to epithelial cells. Understanding the mechanisms of ELT could lead to novel therapeutic strategies for inhibiting tumor metastasis.

10.
Subcell Biochem ; 75: 221-54, 2014.
Article in English | MEDLINE | ID: mdl-24146382

ABSTRACT

Carbonic anhydrase IX (CAIX) which is a zinc containing metalloprotein, efficiently catalyzes the reversible hydration of carbon dioxide. It is constitutively up-regulated in several cancer types and has an important role in tumor progression, acidification and metastasis. High expression of CAIX generally correlates with poor prognosis and is related to a decrease in the disease-free interval following successful therapy. Therefore, it is considered as a prognostic indicator in oncology.In this review, we describe CAIX regulation and its role in tumor hypoxia, acidification and metastasis. In addition, the molecular imaging of CAIX and its potential for use in cancer detection, diagnosis, staging, and for use in following therapy response is discussed. Both antibodies and small molecular weight compounds have been used for targeted imaging of CAIX expression. The use of CAIX expression as an attractive and promising candidate marker for systemic anticancer therapy is also discussed.


Subject(s)
Antigens, Neoplasm/genetics , Carbonic Anhydrases/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Neoplasms/enzymology , Antigens, Neoplasm/biosynthesis , Carbon Dioxide/metabolism , Carbonic Anhydrase IX , Carbonic Anhydrases/biosynthesis , Cell Hypoxia/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Zinc/metabolism
11.
Bioorg Med Chem ; 21(17): 5029-38, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23890524

ABSTRACT

Probes for use in time-resolved fluorescence competitive binding assays at melanocortin receptors based on the parental ligands MSH(4), MSH(7), and NDP-α-MSH were prepared by solid phase synthesis methods, purified, and characterized. The saturation binding of these probes was studied using HEK-293 cells engineered to overexpress the human melanocortin 4 receptor (hMC4R) as well as the human cholecystokinin 2 receptor (hCCK2R). The ratios of non-specific binding to total binding approached unity at high concentrations for each probe. At low probe concentrations, receptor-mediated binding and uptake was discernable, and so probe concentrations were kept as low as possible in determining Kd values. The Eu-DTPA-PEGO-MSH(4) probe exhibited low specific binding relative to non-specific binding, even at low nanomolar concentrations, and was deemed unsuitable for use in competition binding assays. The Eu-DTPA-PEGO probes based on MSH(7) and NDP-α-MSH exhibited Kd values of 27±3.9nM and 4.2±0.48nM, respectively, for binding with hMC4R. These probes were employed in competitive binding assays to characterize the interactions of hMC4R with monovalent and divalent MSH(4), MSH(7), and NDP-α-MSH constructs derived from squalene. Results from assays with both probes reflected only statistical enhancements, suggesting improper ligand spacing on the squalene scaffold for the divalent constructs. The Ki values from competitive binding assays that employed the MSH(7)-based probe were generally lower than the Ki values obtained when the probe based on NDP-α-MSH was employed, which is consistent with the greater potency of the latter probe. The probe based on MSH(7) was also competed with monovalent, divalent, and trivalent MSH(4) constructs that previously demonstrated multivalent binding in competitive binding assays against a variant of the probe based on NDP-α-MSH. Results from these assays confirm multivalent binding, but suggest a more modest increase in avidity for these MSH(4) constructs than was previously reported.


Subject(s)
Fluorescent Dyes/chemical synthesis , Receptors, Melanocortin/metabolism , Binding, Competitive , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , HEK293 Cells , Humans , Pentetic Acid/chemistry , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Protein Binding , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Melanocortin/chemistry , Solid-Phase Synthesis Techniques
12.
Mol Pharm ; 10(8): 3175-85, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23763620

ABSTRACT

The melanocortin 1 receptor (MC1R) is overexpressed in most melanoma metastases, making it a promising target for imaging of melanomas. In this study, the expression of MC1R in a large fraction of patients with melanoma was confirmed using mRNA and tissue microarray. Here, we have characterized the in vivo tumor and tissue distribution and pharmacokinetics (PK) of uptake and clearance of a MC1R specific peptidomimetic ligand conjugated to a near-infrared fluorescent dye. We propose an interdisciplinary framework to bridge the different time and space scales of ligand-tumor-host interactions: intravital fluorescence microscopy to quantify probe internalization at the cellular level, a xenograft tumor model for whole body pharmacokinetics, and a computational pharmacokinetic model for integration and interpretation of experimental data. Administration of the probe into mice bearing tumors with high and low MC1R expression demonstrated normalized image intensities that correlated with expression levels (p < 0.05). The biodistribution study showed high kidney uptake as early as 30 min postinjection. The PK computational model predicted the presence of receptors in the kidneys with a lower affinity, but at higher numbers than in the tumors. As the mouse kidney is known to express the MC5R, this hypothesis was confirmed by both coinjection of a ligand with higher MC5R affinity compared to MC1R and by injection of lower probe concentrations (e.g., 1 nmol/kg), both leading to decreased kidney accumulation of the MC1R ligand. In addition, through this interdisciplinary approach we could predict the rates of ligand accumulation and clearance into and from organs and tumors, and the amount of injected ligand required to have maximum specific retention in tumors. These predictions have potential to aid in the translation of a targeted agent from lab to the clinic. In conclusion, the characterized MC1R-specific probe has excellent potential for in vivo detection of melanoma metastases. The process of cell-surface marker validation, targeted imaging probe development, and in vitro, in vivo, and in silico characterization described in this study can be generally applied to preclinical development of targeted agents.


Subject(s)
Melanoma/metabolism , Molecular Probes/metabolism , Molecular Probes/pharmacokinetics , Receptor, Melanocortin, Type 1/metabolism , Animals , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Mice , Models, Theoretical , Receptor, Melanocortin, Type 1/genetics , Tissue Array Analysis , Xenograft Model Antitumor Assays
13.
Bioconjug Chem ; 23(12): 2451-9, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23116461

ABSTRACT

The incidence of malignant melanoma is rising more rapidly than that of any other cancer in the United States. The melanocortin 1 receptor (MC1R) is overexpressed in most human melanoma metastases, thus making it a promising target for imaging and therapy of melanomas. We have previously reported the development of a peptidomimetic ligand with high specificity and affinity for MC1R. Here, we have conjugated near-infrared fluorescent dyes to the C-terminus of this ligand via lysine-mercaptopropionic acid linkers to generate MC1R specific optical probes (MC1RL-800, 0.4 nM K(i); and MC1RL-Cy5, 0.3 nM K(i)). Internalization of the imaging probe was studied in vitro by fluorescence microscopy using engineered A375/MC1R cells and B16F10 cells with endogenous MC1R expression. The in vivo tumor targeting of MC1RL-800 was evaluated by intravenous injection of probe into nude mice bearing bilateral subcutaneous A375 xenograft tumors with low MC1R expression and engineered A375/MC1R tumors with high receptor expression. Melanotic B16F10 xenografts were also studied. Fluorescence imaging showed that the agent has higher uptake values in tumors with high expression compared to low (p < 0.05), demonstrating the effect of expression levels on image contrast-to-noise. In addition, tumor uptake was significantly blocked by coinjection of excess NDP-α-MSH peptide (p < 0.05). In conclusion, the MC1R-specific imaging probe developed in this study displays excellent potential for the intraoperative detection of regional node involvement and for margin detection during melanoma metastasis resection.


Subject(s)
Fluorescent Dyes/chemical synthesis , Melanoma, Experimental/pathology , Neoplasm Proteins/analysis , Peptidomimetics/chemical synthesis , Receptor, Melanocortin, Type 1/analysis , Skin Neoplasms/pathology , Animals , Endocytosis/drug effects , Fluorescent Dyes/metabolism , Gene Expression , Humans , Injections, Intravenous , Ligands , Male , Melanoma, Experimental/diagnosis , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Microscopy, Fluorescence , Molecular Imaging/methods , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Peptidomimetics/metabolism , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
15.
Clin Cancer Res ; 18(1): 207-19, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22016510

ABSTRACT

PURPOSE: To develop targeted molecular imaging probes for the noninvasive detection of breast cancer lymph node metastasis. EXPERIMENTAL DESIGN: Six cell surface or secreted markers were identified by expression profiling and from the literature as being highly expressed in breast cancer lymph node metastases. Two of these markers were cell surface carbonic anhydrase isozymes (CAIX and/or CAXII) and were validated for protein expression by immunohistochemistry of patient tissue samples on a breast cancer tissue microarray containing 47 normal breast tissue samples, 42 ductal carcinoma in situ, 43 invasive ductal carcinomas without metastasis, 46 invasive ductal carcinomas with metastasis, and 49 lymph node macrometastases of breast carcinoma. Targeted probes were developed by conjugation of CAIX- and CAXII-specific monoclonal antibodies to a near-infrared fluorescent dye. RESULTS: Together, these two markers were expressed in 100% of the lymph node metastases surveyed. Selectivity of the imaging probes were confirmed by intravenous injection into nude mice-bearing mammary fat pad tumors of marker-expressing cells and nonexpressing cells or by preinjection of unlabeled antibody. Imaging of lymph node metastases showed that peritumorally injected probes detected nodes harboring metastatic tumor cells. As few as 1,000 cells were detected, as determined by implanting, under ultrasound guidance, a range in number of CAIX- and CAXII-expressing cells into the axillary lymph nodes. CONCLUSION: These imaging probes have potential for noninvasive staging of breast cancer in the clinic and elimination of unneeded surgery, which is costly and associated with morbidities.


Subject(s)
Antibodies, Monoclonal , Antigens, Neoplasm/metabolism , Breast Neoplasms/diagnosis , Carbonic Anhydrases/metabolism , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Diagnostic Imaging , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Breast/immunology , Breast/metabolism , Breast/pathology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Carbonic Anhydrase IX , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/immunology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Luciferases/metabolism , Luminescent Measurements , Lymphatic Metastasis , Mice , Mice, Nude , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Tissue Distribution , Tumor Cells, Cultured
16.
J Med Chem ; 54(23): 8078-84, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22011200

ABSTRACT

The incidence of malignant melanoma is rising faster than that of any other cancer in the United States. Because of its high expression on the surface of melanomas, MC1R has been investigated as a target for selective imaging and therapeutic agents against melanoma. Eight ligands were screened against cell lines engineered to overexpress MC1R, MC4R, or MC5R. Of these, compound 1 (4-phenylbutyryl-His-dPhe-Arg-Trp-NH(2)) exhibited high (0.2 nM) binding affinity for MC1R and low (high nanomolar) affinities for MC4R and MC5R. Functionalization of the ligand at the C-terminus with an alkyne for use in Cu-catalyzed click chemistry was shown not to affect the binding affinity. Finally, formation of the targeted polymer, as well as the targeted micelle formulation, also resulted in constructs with low nanomolar binding affinity.


Subject(s)
Antineoplastic Agents/chemical synthesis , Azides/chemistry , Oligopeptides/chemical synthesis , Polyethylene Glycols/chemistry , Proteins/chemistry , Receptor, Melanocortin, Type 1/metabolism , Alkynes/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding, Competitive , Click Chemistry , Drug Screening Assays, Antitumor , HCT116 Cells , HEK293 Cells , Humans , Ligands , Melanoma , Micelles , Oligopeptides/chemistry , Oligopeptides/pharmacology , Receptor, Melanocortin, Type 4/metabolism , Receptors, Melanocortin/metabolism , Structure-Activity Relationship
17.
Bioorg Med Chem ; 19(21): 6474-82, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21940174

ABSTRACT

A spherical molecular scaffold bearing eight terminal alkyne groups was synthesized in one step from sucrose. One or more copies of a tetrapeptide azide, either N(3)(CH(2))(5)(CO)-His-DPhe-Arg-Trp-NH(2) (MSH4) or N(3)(CH(2))(5)(CO)-Trp-Met-Asp-Phe-NH(2) (CCK4), were attached to the scaffold via the copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) reaction. Competitive binding assays using Eu-labeled probes based on the superpotent ligands Ser-Tyr-Ser-Nle-Glu-His-DPhe-Arg-Trp-Gly-Lys-Pro-Val-NH(2) (NDP-α-MSH) and Asp-Tyr-Met-Gly-Trp-Met-Asp-Phe-NH(2) (CCK8) were used to study the interactions of monovalent and multivalent MSH4 and CCK4 constructs with Hek293 cells engineered to overexpress MC4R and CCK2R. All of the monovalent and multivalent MSH4 constructs exhibited binding comparable to that of the parental ligand, suggesting that either the ligand spacing was inappropriate for multivalent binding, or MSH4 is too weak a binder for a second 'anchoring' binding event to occur before the monovalently-bound construct is released from the cell surface. In contrast with this behavior, monovalent CCK4 constructs were significantly less potent than the parental ligand, while multivalent CCK4 constructs were as or more potent than the parental ligand. These results are suggestive of multivalent binding, which may be due to increased residence times for monovalently bound CCK4 constructs on the cell surface relative to MSH4 constructs, the greater residence time being necessary for the establishment of multivalent binding.


Subject(s)
Alkynes/chemistry , Cell Cycle Proteins/chemistry , Protein Multimerization , Solid-Phase Synthesis Techniques/methods , Sucrose/chemistry , Tetragastrin/chemistry , Binding, Competitive , Cell Cycle Proteins/metabolism , Cell Line , Magnetic Resonance Spectroscopy , Molecular Structure , Regression Analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tetragastrin/metabolism
18.
Cancer Res ; 71(3): 1050-9, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21169406

ABSTRACT

Pathologic axillary lymph node (ALN) status is an important prognostic factor for staging breast cancer. Currently, status is determined by histopathology following surgical excision of sentinel lymph node(s), which is an invasive, time consuming, and costly procedure with potential morbidity to the patient. Here, we describe an imaging platform for noninvasive assessment of ALN status, eliminating the need for surgical examination of patients to rule out nodal involvement. A targeted imaging probe (MamAb-680) was developed by conjugation of a mammaglobin-A-specific monoclonal antibody to a near-infrared fluorescent dye. Using DNA and tissue microarray, mammaglobin-A was validated as a cell-surface target that is expressed in ALN-positive patient samples but is not expressed in normal lymph nodes. In vivo selectivity was determined by i.v. injection of MamAb-680 into mice with mammaglobin-A-positive and -negative mammary fat pad (MFP) tumors; and by peritumoral MFP injection of the targeted imaging probe in mice with spontaneous ALN metastases. Fluorescence imaging showed that probe was only retained in positive tumors and metastases. As few as 1,000 cells that endogenously express mammaglobin-A were detected in ALN, indicating high sensitivity of this method. Translation of this approach offers considerable potential as a noninvasive clinical strategy to stage breast cancer.


Subject(s)
Antibodies, Monoclonal/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Immunoconjugates/metabolism , Lymph Nodes/pathology , Neoplasm Proteins/biosynthesis , Uteroglobin/biosynthesis , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Diagnostic Imaging/methods , Female , Fluorescent Dyes/metabolism , Fluorescent Dyes/pharmacokinetics , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphatic Metastasis , Mammaglobin A , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transplantation, Heterologous , Uteroglobin/genetics , Uteroglobin/immunology
19.
Cancer Control ; 17(3): 143-55, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20664511

ABSTRACT

BACKGROUND: Significant efforts have been directed toward developing and enhancing imaging methods for the early detection, diagnosis, and characterization of small breast tumors. Molecular and functional imaging sets the stage for enhancement of current methodology. METHODS: Current imaging modalities are described based on the molecular characteristics of normal and malignant tissue. New molecular imaging methods that have the potential for clinical use are also discussed. RESULTS: Dynamic contrast-enhanced magnetic resonance imaging is more sensitive than mammography in BRCA1 carriers. It is used in screening and in the early evaluation of neoadjuvant therapy. Positron emission mammography is 91% sensitive and 93% specific in detecting primary breast cancers. Sentinel node scintigraphy is a key component of axillary lymph node evaluation. Other imaging modalities being studied include Tc99m sestamibi, radiolabeled thymidine or uridine, estrogen receptor imaging, magnetic resonance spectroscopy, and diffusion magnetic resonance imaging. CONCLUSIONS: Molecular and functional imaging of the breast will likely alter clinical practice in diagnosing and staging primary breast cancer and assessing response to therapy since it will provide earlier information regarding the underlying biology of individual breast cancers, tumor stage, potential treatment strategies, and biomarkers for early evaluation of treatment effects.


Subject(s)
Breast Neoplasms/diagnosis , Diagnostic Imaging/methods , Female , Humans , Magnetic Resonance Imaging/methods , Mammography/methods , Positron-Emission Tomography/methods
20.
Biochem Pharmacol ; 80(5): 748-54, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20510208

ABSTRACT

Pancreatic cancer has a high mortality rate, which is generally related to the initial diagnosis coming at late stage disease combined with a lack of effective treatment options. Novel agents that selectively detect pancreatic cancer have potential for use in the molecular imaging of cancer, allowing for non-invasive determination of tumor therapeutic response and molecular characterization of the disease. Such agents may also be used for the targeted delivery of therapy to tumor cells while decreasing systemic effects. Using complementary assays of mRNA expression profiling to determine elevated expression in pancreatic cancer tissues relative to normal pancreas tissues, and validation of protein expression by immunohistochemistry on tissue microarray, we have identified cell-surface targets with potential for imaging and therapeutic agent development. Expression profiles of 2177 cell-surface genes for 28 pancreatic tumor specimens and 4 normal pancreas tissue samples were evaluated. Expression in normal tissues was evaluated using array data from 103 samples representing 28 organ sites as well as mining published data. One-hundred seventy unique targets were highly expressed in 2 or more of the pancreatic tumor specimens and were not expressed in the normal pancreas samples. Two targets (TLR2 and ABCC3) were further validated for protein expression by tissue microarray (TMA) based immunohistochemistry. These validated targets have potential for the development of diagnostic imaging and therapeutic agents for pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Gene Expression Profiling , Pancreatic Neoplasms/drug therapy , Tissue Array Analysis , Adenocarcinoma/genetics , Humans , Immunohistochemistry , Pancreatic Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...