Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38324296

ABSTRACT

PURPOSE: Antibody-drug conjugates (ADCs) have shown impressive clinical activity with approval of many agents in hematological and solid tumors. However, challenges remain with both efficacy and safety of ADCs. This study describes novel trastuzumab-auristatin conjugates with the hydrophilic MMAE prodrug MMAU, and optimization of a glycopeptide linker leading to a wider therapeutic window. EXPERIMENTAL DESIGN: Trastuzumab was conjugated with auristatin payloads via a series of linkers using a stabilized maleimide handle. The ADCs were characterized in vitro and their relative in vivo anti-tumor efficacies were assessed in HER2+ xenograft models. Relative linker stabilities and the mechanism of linker cleavage were studied using in vitro assays. Toxicity and toxicokinetics of the best performing ADC were evaluated in cynomolgus monkey (cyno). RESULTS: The trastuzumab-MMAU ADC with stabilized glycopeptide linker showed maleimide stabilization and higher resistance to cleavage by serum and lysosomal enzymes compared to a valine-citrulline conjugated trastuzumab ADC (trastuzumab-vc-MMAE). A single dose of 1 or 2 mg/kg of trastuzumab-MMAU at drug-to-antibody ratios (DAR) of 8 and 4 respectively resulted in xenograft tumor growth inhibition, with superior efficacy to trastuzumab-vc-MMAE. Trastuzumab-MMAU DAR4 was tolerated at doses up to 12 mg/kg in cyno, which represents 2- to 4-fold higher dose than that observed with vedotin ADCs, and had increased terminal half-life and exposure. CONCLUSIONS: The optimized trastuzumab-MMAU ADC showed potent antitumor activity and was well tolerated with excellent pharmacokinetics in non-human primates, leading to a superior preclinical therapeutic window. The data supports potential utility of trastuzumab-MMAU for treatment of HER2+ tumors.

2.
NPJ Breast Cancer ; 8(1): 41, 2022 Mar 24.
Article in English | MEDLINE | ID: mdl-35332139

ABSTRACT

The staging and local management of breast cancer involves the evaluation of the extent and completeness of excision of both the invasive carcinoma component and also the intraductal component or ductal carcinoma in situ. When both invasive ductal carcinoma and coincident ductal carcinoma in situ are present, assessment of the extent and localization of both components is required for optimal therapeutic planning. We have used a mouse model of breast cancer to evaluate the feasibility of applying molecular imaging to assess the local status of cancers in vivo. Multi-tracer positron emission tomography (PET) and magnetic resonance imaging (MRI) characterize the transition from premalignancy to invasive carcinoma. PET tracers for glucose consumption, membrane synthesis, and neoangiogenesis in combination with a Gaussian mixture model-based analysis reveal image-derived thresholds to separate the different stages within the whole-lesion. Autoradiography, histology, and quantitative image analysis of immunohistochemistry further corroborate our in vivo findings. Finally, clinical data further support our conclusions and demonstrate translational potential. In summary, this preclinical model provides a platform for characterizing multistep tumor progression and provides proof of concept that supports the utilization of advanced protocols for PET/MRI in clinical breast cancer imaging.

3.
Toxicol Pathol ; 49(3): 505-520, 2021 04.
Article in English | MEDLINE | ID: mdl-33380270

ABSTRACT

Normal retina and its cell layers are essential for processing visual stimuli, and loss of its integrity has been documented in many disease processes. The numbers and the axonal processes of retinal ganglion cells are reduced substantially in glaucoma, leading to vision loss and blindness. Similarly, selective loss of photoreceptors in age-related macular degeneration and hereditary retinal dystrophies also results in the compromise of visual acuity. Development of genetically modified mice has led to increased understanding of the pathogenesis of many retinal diseases. Similarly, in this digital era, usage of modalities to quantify the retinal cell loss has grown exponentially leading to a better understanding of the suitability of animal models to study human retinal diseases. These quantification modalities provide valuable quantifiable data in studying pathogenesis and disease progression. This review will discuss the immunohistochemical markers for various retinal cells, available automated tools to quantify retinal cells, and present an example of retinal ganglion cell quantification using HALO image analysis platform. Additionally, we briefly review retinal cell types and subtypes, salient features of retina in various laboratory animal species, and a few of the main disease processes that affect retinal cell numbers in humans.


Subject(s)
Retina , Retinal Ganglion Cells , Animals , Axons , Disease Models, Animal , Humans , Mice , Photoreceptor Cells
4.
Cancer Immunol Res ; 8(5): 596-608, 2020 05.
Article in English | MEDLINE | ID: mdl-32184296

ABSTRACT

Patients with hematologic cancers have improved outcomes after treatment with bispecific antibodies that bind to CD3 on T cells and that redirect T cells toward cancer cells. However, clinical benefit against solid tumors remains to be shown. We made a bispecific antibody that targets both the common prostate tumor-specific antigen PSMA and CD3 (PMSAxCD3) and provide evidence for tumor inhibition in several preclinical solid tumor models. Mice expressing the human extracellular regions of CD3 and PSMA were generated to examine antitumor efficacy in the presence of an intact immune system and PSMA expression in normal tissues. PSMAxCD3 accumulated in PSMA-expressing tissues and tumors as detected by immuno-PET imaging. Although PSMAxCD3 induced T-cell activation and showed antitumor efficacy in mice with low tumor burden, PSMAxCD3 lost efficacy against larger solid tumors, mirroring the difficulty of treating solid tumors in the clinic. Costimulatory receptors can enhance T-cell responses. We show here that costimulation can enhance the antitumor efficacy of PSMAxCD3. In particular, 4-1BB stimulation in combination with PSMAxCD3 enhanced T-cell activation and proliferation, boosted efficacy against larger tumors, and induced T-cell memory, leading to durable antitumor responses. The combination of CD3 bispecific antibodies and anti-4-1BB costimulation represents a therapeutic approach for the treatment of solid tumors.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , CD3 Complex/immunology , Glutamate Carboxypeptidase II/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Animals , Antibodies, Bispecific/immunology , Antigens, Surface/immunology , Antigens, Surface/metabolism , CD3 Complex/metabolism , Cell Line, Tumor , Disease Models, Animal , Glutamate Carboxypeptidase II/immunology , Glutamate Carboxypeptidase II/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Zirconium/pharmacokinetics
5.
Toxicol Pathol ; 48(2): 277-294, 2020 02.
Article in English | MEDLINE | ID: mdl-31645203

ABSTRACT

Toxicologic pathology is transitioning from analog to digital methods. This transition seems inevitable due to a host of ongoing social and medical technological forces. Of these, artificial intelligence (AI) and in particular machine learning (ML) are globally disruptive, rapidly growing sectors of technology whose impact on the long-established field of histopathology is quickly being realized. The development of increasing numbers of algorithms, peering ever deeper into the histopathological space, has demonstrated to the scientific community that AI pathology platforms are now poised to truly impact the future of precision and personalized medicine. However, as with all great technological advances, there are implementation and adoption challenges. This review aims to define common and relevant AI and ML terminology, describe data generation and interpretation, outline current and potential future business cases, discuss validation and regulatory hurdles, and most importantly, propose how overcoming the challenges of this burgeoning technology may shape toxicologic pathology for years to come, enabling pathologists to contribute even more effectively to answering scientific questions and solving global health issues. [Box: see text].


Subject(s)
Artificial Intelligence , Pathology/methods , Toxicology/methods , Humans , Image Processing, Computer-Assisted/methods
6.
Proc Natl Acad Sci U S A ; 114(5): E707-E716, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096336

ABSTRACT

Aberrant activation of ß-catenin through its activity as a transcription factor has been observed in a large proportion of human malignancies. Despite the improved understanding of the ß-catenin signaling pathway over the past three decades, attempts to develop therapies targeting ß-catenin remain challenging, and none of these targeted therapies have advanced to the clinic. In this study, we show that part of the challenge in antagonizing ß-catenin is caused by its dual functionality as a cell adhesion molecule and a signaling molecule. In a mouse model of basal ErbB2 receptor tyrosine kinase 2 (ErbB2)-positive breast cancer (ErbB2KI), which exhibits aberrant ß-catenin nuclear signaling, ß-catenin haploinsufficiency induced aggressive tumor formation and metastasis by promoting the disruption of adherens junctions, dedifferentiation, and an epithelial to mesenchymal transition (EMT) transcriptional program. In contrast to the accelerated tumor onset observed in the haploid-insufficient ErbB2 tumors, deletion of both ß-catenin alleles in the ErbB2KI model had only a minor impact on tumor onset that further correlated with the retention of normal adherens junctions. We further showed that retention of adherens junctional integrity was caused by the up-regulation of the closely related family member plakoglobin (γ-catenin) that maintained both adherens junctions and the activation of Wnt target genes. In contrast to the ErbB2KI basal tumor model, modulation of ß-catenin levels had no appreciable impact on tumor onset in an ErbB2-driven model of luminal breast cancer [murine mammary tumor virus promoter (MMTV-NIC)]. These observations argue that the balance of junctional and nuclear ß-catenin activity has a profound impact on tumor progression in this basal model of ErbB2-positive breast cancer.


Subject(s)
Mammary Neoplasms, Experimental/pathology , Receptor, ErbB-2/metabolism , beta Catenin/genetics , Animals , Epithelial-Mesenchymal Transition , Female , Haploinsufficiency , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice, Transgenic , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Signal Transduction , Tumor Cells, Cultured , gamma Catenin/genetics
7.
Cancer Res ; 76(18): 5512-22, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27466286

ABSTRACT

Differential diagnosis and therapy of heterogeneous breast tumors poses a major clinical challenge. To address the need for a comprehensive, noninvasive strategy to define the molecular and functional profiles of tumors in vivo, we investigated a novel combination of metabolic PET and diffusion-weighted (DW)-MRI in the polyoma virus middle T antigen transgenic mouse model of breast cancer. The implementation of a voxelwise analysis for the clustering of intra- and intertumoral heterogeneity in this model resulted in a multiparametric profile based on [(18)F]Fluorodeoxyglucose ([(18)F]FDG)-PET and DW-MRI, which identified three distinct tumor phenotypes in vivo, including solid acinar, and solid nodular malignancies as well as cystic hyperplasia. To evaluate the feasibility of this approach for clinical use, we examined estrogen receptor-positive and progesterone receptor-positive breast tumors from five patient cases using DW-MRI and [(18)F]FDG-PET in a simultaneous PET/MRI system. The postsurgical in vivo PET/MRI data were correlated to whole-slide histology using the latter traditional diagnostic standard to define phenotype. By this approach, we showed how molecular, structural (microscopic, anatomic), and functional information could be simultaneously obtained noninvasively to identify precancerous and malignant subtypes within heterogeneous tumors. Combined with an automatized analysis, our results suggest that multiparametric molecular and functional imaging may be capable of providing comprehensive tumor profiling for noninvasive cancer diagnostics. Cancer Res; 76(18); 5512-22. ©2016 AACR.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Diffusion Magnetic Resonance Imaging/methods , Image Interpretation, Computer-Assisted/methods , Multimodal Imaging/methods , Positron-Emission Tomography/methods , Aged , Animals , Disease Models, Animal , Female , Fluorodeoxyglucose F18 , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Radiopharmaceuticals
8.
Mol Carcinog ; 55(9): 1387-96, 2016 09.
Article in English | MEDLINE | ID: mdl-26310697

ABSTRACT

The tumor suppressor TP53 can initiate a plethora of anti-proliferative effects to maintain genomic integrity under conditions of genotoxic stress. The N-terminal proline-rich domain (PRD) of TP53 is important in the regulation of TP53 activity and stability. A common polymorphism at codon 72 in this region has been associated with altered cancer risk in humans. The Trp53ΔP mouse, which carries a germline homozygous deletion of a region of the PRD, does not develop spontaneous tumors in a mixed 129/Sv and C57BL/6 genetic background, but is highly susceptible to a broad range of tumor types following total body exposure to 4 Gy gamma (γ) radiation. This contrasts with the tumor spectrum in Trp53 null (-/-) mice, which mainly develop thymic lymphomas and osteosarcomas. Analysis of genomic instability in tissues and cells from Trp53ΔP mice demonstrated elevated basal levels of aneuploidy, but this is not sufficient to drive spontaneous tumorigenesis, which requires an additional DNA damage stimulus. Levels of genomic instability did not increase significantly in Trp53ΔP mice following irradiation exposure, suggesting that other radiation effects including tissue inflammation, altered metabolism or autophagy, may play an important role. The Trp53ΔP mouse is a novel model to dissect the mechanisms of tumor development induced by radiation exposure. © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinogenesis/genetics , Genomic Instability , Neoplasms, Radiation-Induced/genetics , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Animals , Autophagy , Female , Gamma Rays , Male , Mice , Mice, Inbred C57BL , Polyploidy , Proline/chemistry , Proline/genetics , Sequence Deletion , Tumor Suppressor Protein p53/chemistry
9.
Nature ; 525(7567): 114-8, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26266975

ABSTRACT

The adult mouse mammary epithelium contains self-sustained cell lineages that form the inner luminal and outer basal cell layers, with stem and progenitor cells contributing to its proliferative and regenerative potential. A key issue in breast cancer biology is the effect of genomic lesions in specific mammary cell lineages on tumour heterogeneity and progression. The impact of transforming events on fate conversion in cancer cells of origin and thus their contribution to tumour heterogeneity remains largely elusive. Using in situ genetic lineage tracing and limiting dilution transplantation, we have unravelled the potential of PIK3CA(H1047R), one of the most frequent mutations occurring in human breast cancer, to induce multipotency during tumorigenesis in the mammary gland. Here we show that expression of PIK3CA(H1047R) in lineage-committed basal Lgr5-positive and luminal keratin-8-positive cells of the adult mouse mammary gland evokes cell dedifferentiation into a multipotent stem-like state, suggesting this to be a mechanism involved in the formation of heterogeneous, multi-lineage mammary tumours. Moreover, we show that the tumour cell of origin influences the frequency of malignant mammary tumours. Our results define a key effect of PIK3CA(H1047R) on mammary cell fate in the pre-neoplastic mammary gland and show that the cell of origin of PIK3CA(H1047R) tumours dictates their malignancy, thus revealing a mechanism underlying tumour heterogeneity and aggressiveness.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Lineage/genetics , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Multipotent Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/genetics , Animals , Cell Dedifferentiation/genetics , Cell Transformation, Neoplastic/genetics , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Multipotent Stem Cells/pathology , Mutation/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/metabolism
10.
PLoS One ; 10(3): e0120037, 2015.
Article in English | MEDLINE | ID: mdl-25785992

ABSTRACT

Under magnetic resonance (MR) guidance, high intensity focused ultrasound (HIFU) is capable of precise and accurate delivery of thermal dose to tissues. Given the excellent soft tissue imaging capabilities of MRI, but the lack of data on the correlation of MRI findings to histology following HIFU, we sought to examine tumor response to HIFU ablation to determine whether there was a correlation between histological findings and common MR imaging protocols in the assessment of the extent of thermal damage. Female FVB mice (n = 34), bearing bilateral neu deletion tumors, were unilaterally insonated under MR guidance, with the contralateral tumor as a control. Between one and five spots (focal size 0.5 × 0.5 × 2.5 mm3) were insonated per tumor with each spot receiving approximately 74.2 J of acoustic energy over a period of 7 seconds. Animals were then imaged on a 7T MR scanner with several protocols. T1 weighted images (with and without gadolinium contrast) were collected in addition to a series of T2 weighted and diffusion weighted images (for later reconstruction into T2 and apparent diffusion coefficient maps), immediately following ablation and at 6, 24, and 48 hours post treatment. Animals were sacrificed at each time point and both insonated/treated and contralateral tumors removed and stained for NADH-diaphorase, caspase 3, or with hematoxylin and eosin (H&E). We found the area of non-enhancement on contrast enhanced T1 weighted imaging immediately post ablation correlated with the region of tissue receiving a thermal dose CEM43 ≥ 240 min. Moreover, while both tumor T2 and apparent diffusion coefficient values changed from pre-ablation values, contrast enhanced T1 weighted images appeared to be more senstive to changes in tissue viability following HIFU ablation.


Subject(s)
Adenocarcinoma/diagnosis , Diffusion Magnetic Resonance Imaging/methods , High-Intensity Focused Ultrasound Ablation/methods , Image Processing, Computer-Assisted/methods , Mammary Neoplasms, Experimental/diagnosis , Tumor Burden/radiation effects , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Animals , Cell Line, Tumor , Contrast Media/chemistry , Contrast Media/metabolism , Female , Gadolinium/chemistry , Gadolinium/metabolism , High-Intensity Focused Ultrasound Ablation/instrumentation , Histocytochemistry , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/surgery , Mice , Sound , Surgery, Computer-Assisted
11.
Urol Oncol ; 30(5): 635-45, 2012 Sep.
Article in English | MEDLINE | ID: mdl-20888272

ABSTRACT

OBJECTIVES: To develop bladder cancer-specific ligands using a combinatorial chemistry approach. MATERIALS AND METHODS: We performed a high-throughput one-bead one-compound combinatorial chemistry approach to identify ligands that bound to bladder transitional cell carcinoma cells. The whole-cell binding assay allowed successful identification of a few peptides that bound selectively to bladder cancer cells. Single cell suspensions derived from clinical bladder cancer specimens and cell lines were used to determine the binding specificity. Studies with mouse xenografts were performed to determine the in vivo binding and targeting efficiency, specificity, and biodistribution of one of the ligands. RESULTS: One cyclic peptide named PLZ4 (amino acid sequence: cQDGRMGFc) was identified that could selectively bind to bladder cancer cell lines and all of the 5 primary bladder cancer cells from human patients, but not to normal urothelial cells, cell mixtures from normal bladder specimens, fibroblasts, and blood cells. Comparison of PLZ4 binding to cell lines of different cancer origins showed that it was bladder cancer-specific (P < 0.05). PLZ4 could bind to tumor cells treated with urine at pH 6.0, but not to noncancerous cells collected from the urine of 4 patients actively being treated with intravesical Bacillus Calmette-Guerin therapy. In vivo and ex vivo imaging studies showed that PLZ4 linked to Cy5.5 fluorescent dye administered via tail vein injection was specifically taken up in mouse xenografts developed from excised fresh human bladder cancer specimens. Several ligands contain the same DGR motif, but only PLZ4 was bladder cancer-specific. We performed alanine walk and rainbow bead coding experiments, and found that the C-terminal GF residues were also important for cell binding and modulated the binding specificity. CONCLUSIONS: PLZ4 has the potential to be used for targeted therapy and imaging detection during diagnosis and follow-up/surveillance of noninvasive and advanced bladder cancer.


Subject(s)
Carcinoma, Transitional Cell/metabolism , Combinatorial Chemistry Techniques/methods , Peptides/metabolism , Urinary Bladder Neoplasms/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Carbocyanines/chemistry , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor , Female , Humans , Jurkat Cells , Ligands , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Peptide Library , Peptides/chemistry , Peptides/pharmacokinetics , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacokinetics , Protein Binding , Tissue Distribution , Transplantation, Heterologous , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology
12.
Vet Immunol Immunopathol ; 143(1-2): 11-9, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21722969

ABSTRACT

There is a need to develop novel targeted imaging and therapeutic agents that can aid in early diagnosis, detection of metastasis and treatment of melanoma. Alpha-3 integrin is overexpressed in 82% of metastatic melanomas in humans and may be a potential target for peptide ligands carrying therapeutic agents. Five melanoma cell lines were generated from canine primary oral and metastatic canine tumors, grown in mice, and validated with melanoma markers Melan A, S-100, Micropthalmia transcription factor (MITF), Tyrosinase, and MART-1. The melanoma cell lines were tested for binding affinity to previously published alpha-3 integrin-binding peptides containing the cdGXGXXc motif. Fluorescent conjugates of the alpha-3 integrin binding OA02 peptide were used to quantify receptor affinity in the cell lines, a specimen of canine primary oral melanoma, and melanoma xenografts. Alpha-3 integrin was expressed by all 5 canine melanoma cell lines. Four of the 5 lines as well as the primary canine tumor showed affinity to alpha-3 integrin binding peptides with the cdGXGXXc motif. Optical imaging of canine melanoma xenografts in nude mice indicates rapid, strong uptake of the optical tracer in the tumor with an average persistence of approximately 48 h. Ex vivo images showed high tumor-to-background ratio, with tumor signals more than twice that of the kidney and other vital organs. We propose that integrin alpha-3 integrin binding ligands could potentially become useful probes for imaging and delivery of cytotoxic agents for the treatment of melanoma.


Subject(s)
Dog Diseases/metabolism , Integrin alpha3/metabolism , Melanoma/veterinary , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Dogs , Fluorescent Dyes , Humans , Ligands , Melanoma/metabolism , Melanoma, Amelanotic/metabolism , Melanoma, Amelanotic/veterinary , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Mouth Neoplasms/veterinary , Translational Research, Biomedical , Transplantation, Heterologous
13.
J Med Chem ; 52(1): 14-9, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-19072684

ABSTRACT

Current cancer chemotherapeutic agents clinically deployed today are designed to be indiscriminately cytotoxic, however, achieving selective targeting of cancer malignancies would allow for improved diagnostic and chemotherapeutic tools. Integrin alpha(4)beta(1), a heterodimeric cell surface receptor, is believed to have a low-affinity conformation in resting normal lymphocytes and an activated high-affinity conformation in cancerous cells, specifically T- and B-cell lymphomas. This highly attractive yet poorly understood receptor has been selectively targeted with the bisaryl urea peptidomimetic antagonist 1. However, concerns regarding its preliminary pharmacokinetic (PK) profile provided an impetus to change the pharmacophore from a bisaryl urea to a 2-arylaminobenzothiazole moiety, resulting in an analogue with improved physicochemical properties, solubility, and kidney:tumor ratio while maintaining potency (6; IC(50) = 53 pM). The results presented herein utilized heterocyclic and solid-phase chemistry, cell adhesion assay, and in vivo optical imaging using the cyanine dye Cy5.5 conjugate.


Subject(s)
Benzothiazoles/chemistry , Benzothiazoles/therapeutic use , Integrin alpha4beta1/antagonists & inhibitors , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/pathology , Animals , Benzothiazoles/administration & dosage , Benzothiazoles/chemical synthesis , Cell Adhesion/drug effects , Cell Line, Tumor , Female , Humans , Inhibitory Concentration 50 , Integrin alpha4beta1/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, T-Cell/metabolism , Male , Mice , Mice, Nude , Molecular Structure , Sesquiterpenes , Structure-Activity Relationship , Substrate Specificity , Xenograft Model Antitumor Assays
14.
J Comb Chem ; 10(4): 599-604, 2008.
Article in English | MEDLINE | ID: mdl-18558750

ABSTRACT

We have developed a new color-encoding method that facilitates high-throughput screening of one-bead one-compound (OBOC) combinatorial libraries. Polymer beads displaying chemical compounds or families of compounds are stained with oil-based organic dyes that are used as coding tags. The color dyes do not affect cell binding to the compounds displayed on the surface of the beads. We have applied such rainbow beads in a multiplex manner to discover and profile ligands against cell surface receptors. In the first application, a series of OBOC libraries with different scaffolds or motifs are each color-coded; small samples of each library are then combined and screened concurrently against live cells for cell attachment. Preferred libraries can be rapidly identified and selected for subsequent large-scale screenings for cell surface binding ligands. In a second application, beads with a series of peptide analogues (e.g., alanine scan) are color-coded, combined, and tested for binding against a specific cell line in a single-tissue culture well; the critical residues required for binding can be easily determined. In a third application, ligands reacting against a series of integrins are color-coded and used as a readily applied research tool to determine the integrin profile of any cell type. One major advantage of this straightforward and yet powerful method is that only an ordinary inverted microscope is needed for the analysis, instead of sophisticated (and expensive) fluorescent microscopes or flow cytometers.


Subject(s)
Combinatorial Chemistry Techniques/methods , Drug Evaluation, Preclinical/methods , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/metabolism , Color , Humans , Oligopeptides/chemistry , Peptide Library , Structure-Activity Relationship , Substrate Specificity
15.
Mol Pharm ; 4(5): 631-51, 2007.
Article in English | MEDLINE | ID: mdl-17880166

ABSTRACT

Several monoclonal antibodies that target cell surface receptors have gained approval by the U.S. Food and Drug Administration and are widely used in the treatment of some cancers. These include but are not limited to the anti-CD20 antibody Rituximab, used in lymphoma treatment, as well as anti-HER-2 antibody for breast cancer therapy. The efficacy of this cancer immunotherapy modality is, however, limited by the large size of the antibody (160 kd) and its relatively nonspecific binding to the reticuloendothelial system. This latter property is particularly problematic if the antibody is used as a vehicle to deliver radionuclides, cytotoxic drugs, or toxins to the tumor site. Peptides, peptidomimetic, or small molecules are thus attractive as alternative cell surface targeting agents for cancer imaging and therapy. Cancer cell surface targeting peptides can be derived from known native peptide hormones such as somatostatin and bombesin, or they can be identified through screening combinatorial peptide libraries against unknown cell surface receptor targets. Phage-display peptide library and one-bead one-compound (OBOC) combinatorial library methods have been successfully used to discover peptides that target cancer cells or tumor blood vessel endothelial cells. The phage-display peptide library method, because of its biological nature, can only display l-amino acid peptides. In contrast, the OBOC combinatorial library method allows for bead-surface display of peptides that contain l-amino acids, d-amino acids, unnatural amino acids, or other organic moieties. We have successfully used the OBOC method to discover and optimize ligands against unique cell surface receptors of prostate cancer, T- and B-cell lymphoma, as well as ovarian and lung cancers, and we have used some of these peptides to image xenografts in nude mice with high specificity. Here, we (i) review the literature on the use of phage-display and OBOC combinatorial library methods to discover cancer and tumor blood vessel targeting ligands, and (ii) report on the use of an ovarian cancer targeting ligand, OA02, as an in vivo PET imaging probe in a xenograft model in nude mice.


Subject(s)
Combinatorial Chemistry Techniques/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Peptides/chemistry , Peptides/metabolism , Animals , Databases, Protein , Humans , Ligands , Positron-Emission Tomography
16.
Mol Imaging ; 4(4): 439-47, 2005.
Article in English | MEDLINE | ID: mdl-16285906

ABSTRACT

Through screening of random one-bead one-compound (OBOC) libraries, we previously identified cyclic peptides with the cDGXGXXc motif that bind to alpha3 integrin subunit on ovarian adenocarcinoma cell lines ES-2, SKOV-3, and CaOV-3. We subsequently synthesized two secondary libraries based on this motif and identified new peptides that bound with a higher affinity to these cell lines. One of the peptides identified from the 20% "down-substituted" focused library was the c-dGHCitGPQ-c ("OA02") peptide. The goal of this study was to determine whether this peptide labeled with near-infrared probes could be detected after intravenous injection in ovarian tumor-bearing mice and if it would selectively localize in the tumor. Three different forms of this peptide were synthesized, "OA02"-biotin (noncovalently linked to streptavidin-Cy5.5); "OA02"-Cy5.5 and "OA02"-AlexaFluo 680. Using a KODAK IS2000MM image station, these peptide probes were used at the near-infrared (NIR) spectra to image nude mice bearing ES-2 (alpha3 integrin positive) and Raji (alpha3 integrin negative) xenografts. The peptide probe displayed highly specific tumor uptake within 15 min, which lasted for 70 min for "OA02"-Cy5.5 and "OA02"-AlexaFluo 680 and for 24 hours for "OA02"-biotin-streptavidin-Cy5.5. Some kidney and bladder signal were noted. Prior injection with anti-alpha3 monoclonal antibody blocked the binding of this peptide to the ES-2 tumors.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Integrin alpha3/metabolism , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/metabolism , Peptides/metabolism , Transplantation, Heterologous , Adenocarcinoma/chemistry , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Combinatorial Chemistry Techniques , Female , Humans , Integrin alpha3/isolation & purification , Mice , Mice, Nude , Ovarian Neoplasms/chemistry , Peptides/chemical synthesis , Peptides/isolation & purification , Positron-Emission Tomography , Radiography , Spectroscopy, Near-Infrared , Staining and Labeling
17.
Mol Cancer Ther ; 4(5): 806-13, 2005 May.
Article in English | MEDLINE | ID: mdl-15897245

ABSTRACT

Using "one-bead one-compound" combinatorial chemistry technology, we generated random peptide libraries containing millions of 90 mum TentaGel beads, each with its own unique amino acid sequence. A cyclic random 8-mer library was screened with CAOV-3 (a human ovarian adenocarcinoma cell line) and beads with a unique ligand that bind to the cell surface receptors were coated by one or more layers of cells. These positive beads were isolated, stripped, and microsequenced. Several peptide motifs were identified from these screenings, some of which were novel and unique, e.g., cDGX(4)GX(6)X(7)c. Structure-activity relationship studies of this peptide revealed that the l-aspartate residue at position 2, the two glycines at positions 3 and 5, and the two d-cysteines at the amino and COOH terminus are critical for activity. In addition, a hydrophobic residue was preferred at position X(4), whereas amino acids at positions X(6) and X(7) were more variable. Binding of this peptide to a number of different cancer cell lines and normal cells was also determined and we observed that peptides with this motif bound preferentially to three other human ovarian cancer cell lines (ES-2, SKOV-3, and OVCAR-3) as well as a human glioblastoma cancer cell line (A172). Structural analysis of the peptides using high-resolution nuclear magnetic resonance spectroscopy revealed strong conformational similarity among all peptides with cX(1)GX(4)GX(6)X(7)c motif. Blocking study with a panel of anti-integrin antibodies strongly suggests alpha3 integrin present on these ovarian adenocarcinoma cells is the target receptor for this peptide.


Subject(s)
Cell Adhesion Molecules/isolation & purification , Integrin alpha3/metabolism , Ovarian Neoplasms/chemistry , Peptide Fragments/chemistry , Peptide Library , Adenocarcinoma/chemistry , Adenocarcinoma/metabolism , Cell Adhesion Molecules/physiology , Combinatorial Chemistry Techniques , Female , Glioblastoma/chemistry , Glioblastoma/metabolism , Humans , Integrin alpha3/genetics , Ligands , Ovarian Neoplasms/metabolism , Peptide Fragments/chemical synthesis , Peptide Fragments/physiology , Protein Binding , Structure-Activity Relationship , Tumor Cells, Cultured
18.
Biopolymers ; 66(3): 184-99, 2002.
Article in English | MEDLINE | ID: mdl-12385037

ABSTRACT

Antitumor monoclonal antibodies have shown clinical promise as cancer cell surface targeting agents. More tumor targeting antibodies are likely to be approved by the FDA in the next few years. However, there are two major limitations in antibody-targeted therapy: large size and nonspecific uptake of the antibody molecules by the liver and the reticuloendothelial system. These result in poor tumor penetration of antibody pharmaceuticals and dose-limiting toxicity to the liver and bone marrow. Peptides are excellent alternative targeting agents for human cancers, and they may alleviate some of the problems with antibody targeting. In the last decade, several investigators have successfully used combinatorial library methods to discover cell surface binding peptides that may be useful for cancer targeting. The phage-display library technique and the "one-bead one-compound" combinatorial library method are the two approaches that have been used. Cancer cell surface receptors or endothelial cell surface receptors of the neovasculature are the two popular therapeutic targets for cancer. Results from preclinical studies with some peptides are encouraging in their targeting potential.


Subject(s)
Neoplasms/drug therapy , Peptides/therapeutic use , Amino Acid Sequence , Combinatorial Chemistry Techniques , Gene Targeting , Humans , Ligands , Molecular Sequence Data , Neoplasms/blood supply , Neoplasms/metabolism , Peptide Library , Peptides/chemistry , Protein Array Analysis , Receptors, Cell Surface/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...