Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Sci ; 24(3)2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36768976

ABSTRACT

Near infrared photoimmunotherapy (NIR-PIT) is a newly developed molecular targeted cancer treatment, which selectively kills cancer cells or immune-regulatory cells and induces therapeutic host immune responses by administrating a cancer targeting moiety conjugated with IRdye700. The local exposure to near-infrared (NIR) light causes a photo-induced ligand release reaction, which causes damage to the target cell, resulting in immunogenic cell death (ICD) with little or no side effect to the surrounding normal cells. Moreover, NIR-PIT can generate an immune response in distant metastases and inhibit further cancer attack by combing cancer cells targeting NIR-PIT and immune regulatory cells targeting NIR-PIT or other cancer treatment modalities. Several recent improvements in NIR-PIT have been explored such as catheter-driven NIR light delivery, real-time monitoring of cancer, and the development of new target molecule, leading to NIR-PIT being considered as a promising cancer therapy. In this review, we discuss the progress of NIR-PIT, their mechanism and design strategies for cancer treatment. Furthermore, the overall possible targeting molecules for NIR-PIT with their application for cancer treatment are briefly summarised.


Subject(s)
Neoplasms , Phototherapy , Cell Line, Tumor , Phototherapy/methods , Immunotherapy/methods , Xenograft Model Antitumor Assays , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Infrared Rays , Neoplasms/drug therapy
2.
Int J Mol Sci ; 24(4)2023 Feb 04.
Article in English | MEDLINE | ID: mdl-36834500

ABSTRACT

Traditional immunohistochemistry (IHC) has already become an essential method of diagnosis and therapy in cancer management. However, this antibody-based technique is limited to detecting a single marker per tissue section. Since immunotherapy has revolutionized the antineoplastic therapy, developing new immunohistochemistry strategies to detect multiple markers simultaneously to better understand tumor environment and predict or assess response to immunotherapy is necessary and urgent. Multiplex immunohistochemistry (mIHC)/multiplex immunofluorescence (mIF), such as multiplex chromogenic IHC and multiplex fluorescent immunohistochemistry (mfIHC), is a new and emerging technology to label multiple biomarkers in a single pathological section. The mfIHC shows a higher performance in cancer immunotherapy. This review summarizes the technologies, which are applied for mfIHC, and discusses how they are employed for immunotherapy research.


Subject(s)
Neoplasms , Humans , Fluorescent Antibody Technique , Immunohistochemistry , Biomarkers , Immunotherapy , Biomarkers, Tumor
3.
Int J Mol Sci ; 23(11)2022 May 30.
Article in English | MEDLINE | ID: mdl-35682800

ABSTRACT

Triple-negative breast cancer (TNBC) is a group of heterogeneous and refractory breast cancers with the absence of estrogen receptor (ER), progesterone receptor (PgR) and epidermal growth factor receptor 2 (HER2). Over the past decade, antibody drug conjugates (ADCs) have ushered in a new era of targeting therapy. Since the epidermal growth factor receptor (EGFR) and epithelial cell adhesion molecule (EpCAM) are over expressed on triple-negative breast cancer, we developed novel ADCs by conjugating benzylguanine (BG)-modified monomethyl auristatin E (MMAE) to EpCAM- and EGFR-specific SNAP-tagged single chain antibody fragments (scFvs). Rapid and efficient conjugation was achieved by SNAP-tag technology. The binding and internalization properties of scFv-SNAP fusion proteins were confirmed by flow cytometry and fluorescence microscopy. The dose-dependent cytotoxicity was evaluated in cell lines expressing different levels of EGFR and EpCAM. Both ADCs showed specific cytotoxicity to EGFR or EpCAM positive cell lines via inducing apoptosis at a nanomolar concentration. Our study demonstrated that EGFR specific scFv-425-SNAP-BG-MMAE and EpCAM-specific scFv-EpCAM-SNAP-BG-MMAE could be promising ADCs for the treatment of TNBC.


Subject(s)
Immunoconjugates , Triple Negative Breast Neoplasms , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , ErbB Receptors/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism
4.
Bioconjug Chem ; 29(11): 3586-3594, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30289242

ABSTRACT

Antibody-based diagnostic and therapeutic reagents armed with effector molecules such as dyes and drugs offer hope in the battle against cancer. Several site-specific conjugation methods have been developed to equip antibodies with such effector molecules, but they tend to be expensive and involve multiple reaction steps. The conjugation of two different effector molecules to a single antibody also remains a major challenge. Here we describe a simple, controlled, and robust method for the dual site-specific conjugation of an antibody with two effector molecules in a single-pot reaction using the self-labeling SNAP and CLIP protein tags. We verified the principle of the method by labeling an epidermal growth factor receptor (EGFR)-specific single-chain antibody fragment (scFv-425) simultaneously with IRDye700 and Alexa-Fluor647. This dual-labeled antibody bound to EGFR+ ovarian cancer cell lines and tissue samples with high specificity, and its phototherapeutic efficacy was confirmed by the selective killing of EGFR+ cells in vitro.


Subject(s)
Single-Chain Antibodies/chemistry , Cell Line, Tumor , Coloring Agents/chemistry , ErbB Receptors/immunology , ErbB Receptors/metabolism , Female , Flow Cytometry , Humans , Immunoconjugates/chemistry , Microscopy, Confocal , Ovarian Neoplasms/pathology , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/immunology
5.
Pharm Res ; 34(4): 696-703, 2017 04.
Article in English | MEDLINE | ID: mdl-28074431

ABSTRACT

PURPOSE: Targeted theranostics is an alternative strategy in cancer management that aims to improve cancer detection and treatment simultaneously. This approach combines potent therapeutic and diagnostic agents with the specificity of different cell receptor ligands in one product. The success of antibody drug conjugates (ADCs) in clinical practice has encouraged the development of antibody theranostics conjugates (ATCs). However, the generation of homogeneous and pharmaceutically-acceptable ATCs remains a major challenge. The aim of this study is to detect and eliminate ovarian cancer cells on-demand using an ATC directed to EGFR. METHODS: An ATC with a defined drug-to-antibody ratio was generated by the site-directed conjugation of IRDye®700 to a self-labeling protein (SNAP-tag) fused to an EGFR-specific antibody fragment (scFv-425). RESULTS: In vitro and ex vivo imaging showed that the ATC based on scFv-425 is suitable for the highly specific detection of EGFR+ ovarian cancer cell, human tissues and ascites samples. The construct was also able to eliminate EGFR+ cells and human ascites cells with IC50 values of 45-66 nM and 40-90 nM, respectively. CONCLUSION: Our experiments provide a framework to create a versatile technology platform for the development of ATCs for precise detection and treatment of ovarian cancer cells.


Subject(s)
Apoptosis/drug effects , ErbB Receptors/metabolism , Immunoconjugates/pharmacology , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/drug therapy , Photochemotherapy , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Female , Fluorescent Dyes/chemistry , Humans , Immunoconjugates/chemistry , Immunoglobulin Variable Region/chemistry , Indoles/chemistry , Inhibitory Concentration 50 , Organosilicon Compounds/chemistry , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Single-Chain Antibodies/chemistry , Spectroscopy, Near-Infrared/methods , Theranostic Nanomedicine
6.
Biomacromolecules ; 14(8): 2510-20, 2013 Aug 12.
Article in English | MEDLINE | ID: mdl-23782069

ABSTRACT

Development of effective polymer-based nanocarriers for the successful application in cancer therapy still remains a great challenge in current research. In the present study we present a dendritic polyglycerol-based multifunctional drug immunoconjugate that specifically targets and kills cancer cell lines expressing epidermal growth factor receptor (EGFR). The nanocarrier was provided with a dendritic core as a multifunctional anchoring point, doxorubicin (Doxo) coupled through a pH-sensitive linker, a fluorescence marker, poly(ethylene glycol), as solubilizing and shielding moiety, and a scFv antibody conjugated through the SNAP-Tag technology. The study provides the proof of principle that SNAP-tag technology can be used to generate drug-carrying nanoparticles efficiently modified with single-chain antibodies to specifically target and destroy cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Dendrimers/pharmacology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Animals , Antineoplastic Agents/chemistry , CHO Cells , Cell Line, Tumor , Cell Survival/drug effects , Cricetinae , Cricetulus , Dendrimers/chemistry , Doxorubicin/chemistry , Drug Delivery Systems , Drug Screening Assays, Antitumor , Guanine/analogs & derivatives , Guanine/chemistry , Humans , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Protein Binding , Recombinant Fusion Proteins/chemistry , Single-Chain Antibodies/chemistry
7.
Bioconjug Chem ; 22(12): 2487-95, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-21995499

ABSTRACT

Cancer cells can be killed by photosensitizing agents that induce toxic effects when exposed to nonhazardous light, but this also causes significant damage to surrounding healthy cells. The specificity of photodynamic therapy can be increased by conjugating photosensitizing agents to antibodies and antibody fragments that bind specifically to tumor cell antigens. However, standard conjugation reactions produce heterogeneous products whose targeting specificity and spectroscopic properties can be compromised. In this study, we used an antibody fragment (scFv-425) that binds to the epidermal growth factor receptor (EGFR) as a model to investigate the use of SNAP-tag fusions as an improved conjugation strategy. The scFv-425-SNAP-tag fusion protein allowed the specific conjugation of a chlorin e6 photosensitizer modified with O(6)-benzylguanine, generating a homogeneous product that was delivered specifically to EGFR(+) cancer cells and resulted in significant, tumor cell-specific cytotoxicity. The impact of our results on the development of photodynamic therapy is discussed.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Single-Chain Antibodies/chemistry , Cell Line, Tumor , Chlorophyllides , ErbB Receptors/metabolism , Gene Expression , Guanine/analogs & derivatives , Guanine/chemistry , Humans , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
8.
Pharmaceuticals (Basel) ; 14(4)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917962

ABSTRACT

In the last few decades, antibody-based diagnostic and therapeutic applications have been well established in medicine and have revolutionized cancer managements by improving tumor detection and treatment. Antibodies are unique medical elements due to their powerful properties of being able to recognize specific antigens and their therapeutic mechanisms such as blocking specific pathways, antibody-dependent cellular cytotoxicity, and complement-dependent cytotoxicity. Furthermore, modification techniques have paved the way for improving antibody properties and to develop new classes of antibody-conjugate-based diagnostic and therapeutic agents. These techniques allow arming antibodies with various effector molecules. However, these techniques are utilizing the most frequently used amino acid residues for bioconjugation, such as cysteine and lysine. These bioconjugation approaches generate heterogeneous products with different functional and safety profiles. This is mainly due to the abundance of lysine and cysteine side chains. To overcome these limitations, different site-direct conjugation methods have been applied to arm the antibodies with therapeutic or diagnostics molecules to generate unified antibody conjugates with tailored properties. This review summarizes some of the enzyme-based site-specific conjugation approaches.

9.
J Cancer Res Clin Oncol ; 147(10): 2923-2933, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34185141

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) is characterized by an unfavorable prognosis and missing systemic therapeutic approaches beside chemotherapy. Targeting the immune checkpoint PD-1/PD-L1 showed promising results in breast cancer and especially in TNBC. The extracellular signal-regulated kinase 1/2 (ERK1/2) is an important driver of carcinogenesis. Here, the effect of combined PD-1/PD-L1 and ERK1/2 inhibitor treatment is investigated of cell growth and intracellular impact of breast cancer cell lines. METHODS: The IC50 values of each inhibitor and the effect of combined treatment were determined in three TNBC cell lines of different subtypes and one non-TNBC cell line. Phospho-specific antibodies were used in western blot analyses to investigate an effect on ERK1/2 activation. Expressions of immune modulatory and cell cycle-associated genes were examined by quantitative reverse transcription PCR. RESULTS: Both inhibitors PD-1/PD-L1 and ERK1/2 impeded the proliferation of TNBC to a higher extent than of non-TNBC. By combined treatment, cell lines were inhibited either synergistically or additively. ERK1/2 and S6 phosphorylation were reduced and expressions of c-Fos and FosL were diminished after ERK1/2 inhibitor as single and combined treatment. Between genes involved in immune modulation, IL-8 was upregulated in TNBC cells after combined treatment. CONCLUSION: In conclusion, combination of PD-1/PD-L1 and ERK1/2 inhibitors showed favorable effects for a new therapy strategy, with better results in TNBC cell lines than in non-TNBC cells. The effects have to be validated in models that can reflect the interaction between immune and tumor cells like the situation in the tumor micro-environment.


Subject(s)
Antineoplastic Agents/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Apoptosis , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
10.
Nat Protoc ; 14(11): 3101-3125, 2019 11.
Article in English | MEDLINE | ID: mdl-31605098

ABSTRACT

Antibody-based diagnostic and therapeutic agents play a substantial role in medicine, especially in cancer management. A variety of chemical, genetic and enzymatic site-specific conjugation methods have been developed for equipping antibodies with effector molecules to generate homogeneous antibody conjugates with tailored properties. However, most of these methods are relatively complicated and expensive and require several reaction steps. Self-labeling proteins such as the SNAP-tag are an innovative solution for addressing these challenges. The SNAP-tag is a modified version of the human DNA repair enzyme alkylguanine-DNA alkyltransferase (AGT), which reacts specifically with O(6)-benzylguanine (BG)-modified molecules via irreversible transfer of an alkyl group to a cysteine residue. It provides a simple, controlled and robust site-specific method for labeling antibodies with different synthetic small effector molecules. Fusing a SNAP-tag to recombinant antibodies allows efficient conjugation of BG-containing substrates by autocatalytic, irreversible transfer of the alkyl group to a cysteine residue in the enzyme's active site under physiological conditions and with a 1:1 stoichiometry. This protocol describes how to generate site-specific SNAP-tag single-chain antibody fragment (scFv) conjugates with different types of BG-modified effector molecules. A specific example is included for the design and production of an scFv-photosensitizer conjugate and its characterization as an immuno-theranostic agent. This protocol includes DNA sequences encoding scFV-SNAP-tag fusion proteins and outlines strategies for expression, purification and testing of the resulting scFv-SNAP-tag-based immuno-conjugates. All experiments can be performed by a graduate-level researcher with basic molecular biology skills within an 8-week time frame.


Subject(s)
Immunoconjugates/chemistry , Single-Chain Antibodies/chemistry , Cell Line , Fluorescent Dyes/chemistry , Humans , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Recombinant Proteins/chemistry , Staining and Labeling
11.
Cancers (Basel) ; 11(12)2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31756933

ABSTRACT

:The epidermal growth factor receptor (EGFR) family contains four transmembrane tyrosine kinases (EGFR1/ErbB1, Her2/ErbB2, Her3/ErbB3 and Her4/ErbB4) and 13 secreted polypeptide ligands. EGFRs are overexpressed in many solid tumors, including breast, pancreas, head-and-neck, prostate, ovarian, renal, colon, and non-small-cell lung cancer. Such overexpression produces strong stimulation of downstream signaling pathways, which induce cell growth, cell differentiation, cell cycle progression, angiogenesis, cell motility and blocking of apoptosis.The high expression and/or functional activation of EGFRs correlates with the pathogenesis and progression of several cancers, which make them attractive targets for both diagnosis and therapy. Several approaches have been developed to target these receptors and/or the EGFR modulated effects in cancer cells. Most approaches include the development of anti-EGFRs antibodies and/or small-molecule EGFR inhibitors. This review presents the state-of-the-art and future prospects of targeting EGFRs to treat breast cancer.

12.
Exp Ther Med ; 18(4): 2401-2412, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31555351

ABSTRACT

Breast cancer is the most common cancer in women worldwide. Despite recent developments in breast cancer detection and treatment, 1.38 million women each year are still affected. Breast cancer heterogeneity at the population and single-cell level, complexity and developing different metastases are setting several challenges to develop efficient breast cancer therapies. RNA interference (RNAi) represents an opportunity to silence gene expression and inhibit specific pathways in cancer cells. In order to reap the full advantages of RNAi-based therapy, different pathways that sustain cancer cells growth have been targeted using specific siRNAs. The present study investigated the ability of a set of cytotoxic siRNAs to inhibit growth of breast cancer cells. These siRNAs are targeting eukaryotic elongation factor 2 (EEF2), polo-like kinase 1 (PLK1), G protein-coupled receptor kinase 4 (GRK4) and sphingosine kinase interacting protein (SKIP5). To facilitate their targeted delivery, the human epidermal growth factor receptor-3 (HER3)-specific aptamer A30 was used. The in vitro results described in this work indicate that combining the highly specific HER3 aptamer with cytotoxic siRNAs targeting (EEF2, PLK1, GRK4 and SKIP5) can inhibit its activity and ultimately suppress proliferation of HER3 positive breast cancer cells.

13.
Nat Commun ; 9(1): 1640, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29691410

ABSTRACT

Mechanical allodynia is a major symptom of neuropathic pain whereby innocuous touch evokes severe pain. Here we identify a population of peripheral sensory neurons expressing TrkB that are both necessary and sufficient for producing pain from light touch after nerve injury in mice. Mice in which TrkB-Cre-expressing neurons are ablated are less sensitive to the lightest touch under basal conditions, and fail to develop mechanical allodynia in a model of neuropathic pain. Moreover, selective optogenetic activation of these neurons after nerve injury evokes marked nociceptive behavior. Using a phototherapeutic approach based upon BDNF, the ligand for TrkB, we perform molecule-guided laser ablation of these neurons and achieve long-term retraction of TrkB-positive neurons from the skin and pronounced reversal of mechanical allodynia across multiple types of neuropathic pain. Thus we identify the peripheral neurons which transmit pain from light touch and uncover a novel pharmacological strategy for its treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hyperalgesia/therapy , Laser Therapy , Membrane Glycoproteins/metabolism , Neuralgia/metabolism , Neuralgia/therapy , Protein-Tyrosine Kinases/metabolism , Sensory Receptor Cells/radiation effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Female , Humans , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Ligands , Male , Membrane Glycoproteins/genetics , Mice , Neuralgia/genetics , Neuralgia/physiopathology , Protein-Tyrosine Kinases/genetics , Sensory Receptor Cells/metabolism , Touch/radiation effects
14.
Anticancer Agents Med Chem ; 17(10): 1434-1440, 2017.
Article in English | MEDLINE | ID: mdl-28270070

ABSTRACT

BACKGROUND: Targeted imaging and therapy (theranostics) is a promising approach for the simultaneous improvement of cancer diagnosis, prognosis and management. Therapeutic and imaging reagents are coupled to tumor-targeting molecules such as antibodies, providing a basis for truly personalized medicine. However, the development of antibody-drug conjugates with acceptable pharmaceutical properties is a complex process and several parameters must be optimized, such as the controlled conjugation method and the drug-to-antibody ratio. OBJECTIVE: The major aim of this work is to address fundamental key challenges for the development of versatile technology platform for generating homogenous immunotheranostic reagent. METHOD: We conjugated the theranostics reagent IRDye700dx to a recombinant antibody fusion protein containing a self-labeling protein (SNAP-tag) which provides a unique reaction site. RESULTS: The resulting conjugate was suitable for the imaging of cancer cells expressing the epidermal growth factor receptor and demonstrated potent phototherapeutic and imaging activities against them. CONCLUSION: Here, we describe a simple, rapid and robust site-directed labeling method that can be used to generate homogeneous immunoconjugate with defined pharmacological properties.


Subject(s)
Antibodies/therapeutic use , Neoplasms/drug therapy , Theranostic Nanomedicine , Antibodies/chemistry , Dose-Response Relationship, Drug , ErbB Receptors/analysis , ErbB Receptors/biosynthesis , Humans , Indoles/chemistry , Indoles/therapeutic use , Molecular Structure , Organosilicon Compounds/chemistry , Organosilicon Compounds/therapeutic use , Photochemotherapy , Structure-Activity Relationship
15.
J Cancer Res Clin Oncol ; 142(5): 1003-11, 2016 May.
Article in English | MEDLINE | ID: mdl-26847542

ABSTRACT

PURPOSE: The term "theranostics" represents a new paradigm in medicine especially for cancer treatment. This term was coined by Funkhouser in 2002 and defines a reagent that combines therapeutic and diagnostic properties. It is widely believed that theranostics agents will have considerable impact on healthcare before, during, and after disease by improving cancer prognosis and management simultaneously. Current theranostics approaches still rely on passive tumor targeting strategies, which have scattergun effects and tend to damage both neoplastic and non-neoplastic cells. METHODS: Here we describe a simple, controlled, and efficient method to generate homogeneous photoimmunotheranostics reagents. This method combines molecular optical imaging, photodynamic therapy, and immunotherapy using SNAP-tag technology. SNAP-tag is a derivative of the O(6)-alkylguanine-DNA alkyltransferase (AGT) which has the ability to efficiently conjugate to O(6)-benzylguanine (BG) molecules under physiological conditions depending on its folding pattern. RESULTS: The theranostics agent was able to specifically recognize various epidermal growth factor receptor (EGFR)-expressing skin cancer cell lines using flow cytometry analysis and confocal microscopy and eliminate them at EC50's of 32-55 nM. CONCLUSIONS: These experiments provide a framework for using SNAP-tag technology to generate homogeneous photoimmunotheranostics reagents with unified pharmacokinetic and therapeutic profiles. Furthermore, the reagent generated in this work could be used to simultaneously monitor and suppress the growth of skin squamous carcinoma and melanoma cells expressing EGFR.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/metabolism , Immunotherapy , Melanoma/drug therapy , Photochemotherapy , Photosensitizing Agents/pharmacology , Skin Neoplasms/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Flow Cytometry , Fluorescent Dyes/metabolism , Guanine/analogs & derivatives , Guanine/chemistry , Humans , Indoles/metabolism , Melanoma/metabolism , Melanoma/pathology , Microscopy, Confocal , Organosilicon Compounds/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/chemistry , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured
16.
Oncotarget ; 7(34): 54925-54936, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27448975

ABSTRACT

Triple-negative breast cancer (TNBC) is a heterogeneous disease in which the tumors do not express estrogen receptor (ER), progesterone receptor (PgR) or human epidermal growth factor receptor 2 (HER2). Classical receptor-targeted therapies such as tamoxifen or trastuzumab are therefore unsuitable and combinations of surgery, chemotherapy and/or radiotherapy are required. Photoimmunotheranostics is a minimally invasive approach in which antibodies deliver nontoxic photosensitizers that emit light to facilitate diagnosis and produce cytotoxic reactive oxygen species to induce apoptosis and/or necrosis in cancer cells. We developed a panel of photoimmunotheranostic agents against three TNBC-associated cell surface antigens. Antibodies against epidermal growth factor receptor (EGFR), epithelial cell adhesion molecule (EpCAM) and chondroitin sulfate proteoglycan 4 (CSPG4) were conjugated to the highly potent near-infrared imaging agent/photosensitizer IRDye®700DX phthalocyanine using SNAP-tag technology achieving clear imaging in both breast cancer cell lines and human biopsies and highly potent phototherapeutic activity with IC50values of 62-165 nM against five different cell lines expressing different levels of EGFR, EpCAM and CSPG4. A combination of all three reagents increased the therapeutic activity against TNBC cells by up to 40%.


Subject(s)
Immunoconjugates/therapeutic use , Indoles/therapeutic use , Organosilicon Compounds/therapeutic use , Photochemotherapy/methods , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chondroitin Sulfate Proteoglycans/immunology , Chondroitin Sulfate Proteoglycans/metabolism , Epithelial Cell Adhesion Molecule/immunology , Epithelial Cell Adhesion Molecule/metabolism , ErbB Receptors/immunology , ErbB Receptors/metabolism , Female , Guanine/analogs & derivatives , Guanine/chemistry , Humans , Immunoconjugates/chemistry , Indoles/chemistry , Isoindoles , Light , MCF-7 Cells , Membrane Proteins/immunology , Membrane Proteins/metabolism , Microscopy, Confocal , Organosilicon Compounds/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Recombinant Fusion Proteins/chemistry , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology , Triple Negative Breast Neoplasms/diagnosis , Triple Negative Breast Neoplasms/metabolism
17.
Curr Pharm Des ; 19(30): 5437-42, 2013.
Article in English | MEDLINE | ID: mdl-23431986

ABSTRACT

In the past two decades, immense advances have been achieved in the engineering, production and purifying of recombinant proteins. These proteins are being widely utilized in many fields of biology, biotechnology and medicine, including diagnostic and therapeutic applications. These applications often require the modification or conjugation of these proteins with other molecules. Researchers are spending many efforts to develop and improve the methods of protein modifications. A main challenge they face is derivatizing proteins without affecting their structure and biological function. The conjugation methods available today include random and specific chemical modifications on endogenous amino acids or carbohydrate of the protein of interest. Other methods utilize self-labeling tags as fusion partners to the original protein enabling site-specific conjugation. SNAP-tag is one of the most promising self-labeling tags, which reacts specifically, rapidly and covalently with benzylguanine (BG) derivatives. SNAP-tag fusion proteins have been successfully used for imaging living cells. Recently, several studies have utilized the SNAP technology for generating antibody-based diagnostic and therapeutic tools. We here review these approaches and their possible impact on improving cancer targeting.


Subject(s)
Molecular Imaging/methods , Proteins/chemistry , Proteins/metabolism , Animals , Antibodies , Humans , Molecular Probes , Pharmaceutical Preparations , Protein Binding
18.
Nucleic Acid Ther ; 23(3): 203-12, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23544955

ABSTRACT

Small interfering RNAs (siRNAs) silence gene expression by triggering the sequence-specific degradation of mRNAs, but the targeted delivery of such reagents remains challenging and a significant obstacle to therapeutic applications. One promising approach is the use of RNA aptamers that bind tumor-associated antigens to achieve the delivery of siRNAs to tumor cells displaying specific antigens. Wholly RNA-based constructs are advantageous because they are inexpensive to synthesize and their immunogenicity is low. We therefore joined an aptamer-recognizing alpha V and integrin beta 3 (αvß3) integrin to a siRNA that targets eukaryotic elongation factor 2 and achieved for the first time the targeted delivery of a siRNA to tumor cells expressing αvß3 integrin, causing the inhibition of cell proliferation and the induction of apoptosis specifically in tumor cells. The impact of our results on the development of therapeutic aptamer-siRNA constructs is discussed.


Subject(s)
Aptamers, Nucleotide/genetics , Gene Silencing , Integrin alphaVbeta3/metabolism , Peptide Elongation Factor 2/antagonists & inhibitors , RNA, Small Interfering/metabolism , Apoptosis , Aptamers, Nucleotide/chemical synthesis , Base Sequence , Cell Line, Tumor , Gene Expression , Humans , Integrin alphaVbeta3/genetics , Molecular Sequence Data , Nucleic Acid Conformation , Peptide Elongation Factor 2/genetics , Peptide Elongation Factor 2/metabolism , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL