Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Drug Res (Stuttg) ; 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134030

ABSTRACT

Sepsis, a life-threatening condition triggered by an uncontrolled response to infection, results in a systemic inflammatory response syndrome (SIRS) and the failure of multiple organs leading to multiple organ dysfunction (MODS). In the present study, we investigated the therapeutic potential of tofacitinib (TOFA), an FDA-approved inhibitor of JAK1 and JAK3 against sepsis, using a mouse model induced by cecal ligation puncture (CLP). Swiss albino mice were employed to replicate the CLP-induced sepsis model and were randomly divided into four groups: control, CLP, 150 mg/kg TOFA, and 300 mg/kg TOFA. Six hours after the last TOFA dose, we collected blood and tissue samples from the liver, lungs, kidneys, and spleen for histological analysis. Blood samples were used to assess granulocyte and lymphocyte percentages. Throughout the experiment, we monitored body weight and short-term survival. Our comparative histological analysis revealed that 150 mg/kg TOFA had a protective effect against multiple organ damage. Conversely, the study highlighted the harmful effects of 300 mg/kg TOFA, primarily due to liver and renal toxicity within this group. In summary, our findings demonstrate that tofacitinib at an optimal dose of 150 mg/kg showed promise as a potential therapeutic intervention for sepsis-induced multiple organ failure. However, caution is warranted when considering higher dosages.

2.
Environ Sci Pollut Res Int ; 30(36): 86328-86337, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37402918

ABSTRACT

Biofilms are made up of bacterial colonies and their extracellular polymeric substances (EPS) matrix, which protects the bacteria from adverse environmental conditions. The increasing drug resistivity of pathogenic bacteria is becoming an emergency for developing new antibacterial agents. In this study, we have synthesized the zinc oxide nanoparticles (ZnO NPs) using the leaf extract of Saraca asoca plant, and the antibacterial and antibiofilm activity of green synthesized ZnO NPs was measured against the biofilm-producing bacteria Bacillus subtilis. The disk diffusion data reveals that the zone of inhibition (ZOI) starts at a concentration of 0.5 mg/mL and minimum inhibition concentration (100 µg/mL) and minimum bactericidal concentration (150 µg/mL) values were also evaluated for green synthesized ZnO nanomaterials. Crystal violet test and microscopic examination were used to assess the impact of produced nanoparticles on biofilm development. The findings indicated a nearly 45%, 64%, and 83% suppression of biofilm development at 0.5 × MIC, 0.75 × MIC, and 1 × MIC value, respectively. The biofilm biomass of the preformed or matured biofilms by the ZnO NPs was evaluated to be 68%, 50%, and 33% at concentrations of 0.5 × MIC, 0.75 × MIC, and 1 × MIC which was concentration-dependent. Moreover, flow cytometry results suggest damage to the bacterial cell membrane. The data indicated that the proportion of dead cells increased with NP concentration in comparison to the control. Therefore, it can be concluded that the green synthetic ZnO nanoparticles showed excellent antibacterial and antibiofilm activity against the Bacillus subtilis bacteria that produce biofilms and that they could be a promising substitute agent for the treatment of biofilms and drug-resistant bacteria.


Subject(s)
Metal Nanoparticles , Nanoparticles , Zinc Oxide , Zinc Oxide/pharmacology , Zinc Oxide/chemistry , Nanoparticles/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biofilms , Microbial Sensitivity Tests , Bacillus subtilis , Metal Nanoparticles/chemistry , Plant Extracts/pharmacology
3.
Pharmaceutics ; 14(3)2022 Mar 16.
Article in English | MEDLINE | ID: mdl-35336029

ABSTRACT

The chemokine receptor 4 (CXCR4) is a promising diagnostic and therapeutic target for the management of various cancers. CXCR4 has been utilized in immunotherapy, targeted drug delivery, and endoradiotherapy. Poly(amidoamine) [PAMAM] dendrimers are well-defined polymers with unique properties that have been used in the fabrication of nanomaterials for several biomedical applications. Here, we describe the formulation and pharmacokinetics of generation-5 CXCR4-targeted PAMAM (G5-X4) dendrimers. G5-X4 demonstrated an IC50 of 0.95 nM to CXCR4 against CXCL12-Red in CHO-SNAP-CXCR4 cells. Single-photon computed tomography/computed tomography imaging and biodistribution studies of 111In-labeled G5-X4 showed enhanced uptake in subcutaneous U87 glioblastoma tumors stably expressing CXCR4 with 8.2 ± 2.1, 8.4 ± 0.5, 11.5 ± 0.9, 10.4 ± 2.6, and 8.8 ± 0.5% injected dose per gram of tissue at 1, 3, 24, 48, and 120 h after injection, respectively. Specific accumulation of [111In]G5-X4 in CXCR4-positive tumors was inhibited by the peptidomimetic CXCR4 inhibitor, POL3026. Our results demonstrate that while CXCR4 targeting is beneficial for tumor accumulation at early time points, differences in tumor uptake are diminished over time as passive accumulation takes place. This study further confirms the applicability of PAMAM dendrimers for imaging and therapeutic applications. It also emphasizes careful consideration of image acquisition and/or treatment times when designing dendritic nanoplatforms for tumor targeting.

4.
Biomolecules ; 12(3)2022 03 05.
Article in English | MEDLINE | ID: mdl-35327597

ABSTRACT

We have synthesized a series of 10 new, PSMA-targeted, near-infrared imaging agents intended for use in vivo for fluorescence-guided surgery (FGS). Compounds were synthesized from the commercially available amine-reactive active NHS ester of DyLight800. We altered the linker between the PSMA-targeting urea moiety and the fluorophore with a view to improve the pharmacokinetics. Chemical yields for the conjugates ranged from 51% to 86%. The Ki values ranged from 0.10 to 2.19 nM. Inclusion of an N-bromobenzyl substituent at the ε-amino group of lysine enhanced PSMA+ PIP tumor uptake, as did hydrophilic substituents within the linker. The presence of a polyethylene glycol chain within the linker markedly decreased renal uptake. In particular, DyLight800-10 demonstrated high specific uptake relative to background signal within kidney, confirmed by immunohistochemistry. These compounds may be useful for FGS in prostate, renal or other PSMA-expressing cancers.


Subject(s)
Glutamate Carboxypeptidase II , Prostatic Neoplasms , Cell Line, Tumor , Fluorescent Dyes/chemistry , Humans , Male , Prostate/pathology , Prostatic Neoplasms/pathology , Spectroscopy, Near-Infrared/methods
5.
Clin Cancer Res ; 27(3): 877-888, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33077574

ABSTRACT

PURPOSE: Stabilization of the transcription factor NRF2 through genomic alterations in KEAP1 and NFE2L2 occurs in a quarter of patients with lung adenocarcinoma and a third of patients with lung squamous cell carcinoma. In lung adenocarcinoma, KEAP1 loss often co-occurs with STK11 loss and KRAS-activating alterations. Despite its prevalence, the impact of NRF2 activation on tumor progression and patient outcomes is not fully defined. EXPERIMENTAL DESIGN: We model NRF2 activation, STK11 loss, and KRAS activation in vivo using novel genetically engineered mouse models. Furthermore, we derive a NRF2 activation signature from human non-small cell lung tumors that we use to dissect how these genomic events impact outcomes and immune contexture of participants in the OAK and IMpower131 immunotherapy trials. RESULTS: Our in vivo data reveal roles for NRF2 activation in (i) promoting rapid-onset, multifocal intrabronchiolar carcinomas, leading to lethal pulmonary dysfunction, and (ii) decreasing elevated redox stress in KRAS-mutant, STK11-null tumors. In patients with nonsquamous tumors, the NRF2 signature is negatively prognostic independently of STK11 loss. Patients with lung squamous cell carcinoma with low NRF2 signature survive longer when receiving anti-PD-L1 treatment. CONCLUSIONS: Our in vivo modeling establishes NRF2 activation as a critical oncogenic driver, cooperating with STK11 loss and KRAS activation to promote aggressive lung adenocarcinoma. In patients, oncogenic events alter the tumor immune contexture, possibly having an impact on treatment responses. Importantly, patients with NRF2-activated nonsquamous or squamous tumors have poor prognosis and show limited response to anti-PD-L1 treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , NF-E2-Related Factor 2/metabolism , AMP-Activated Protein Kinase Kinases/genetics , AMP-Activated Protein Kinases/genetics , Animals , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Kaplan-Meier Estimate , Kelch-Like ECH-Associated Protein 1/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , NF-E2-Related Factor 2/genetics , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics
6.
Cell Death Discov ; 6(1): 125, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-33298881

ABSTRACT

Malaria remains a major public health problem worldwide. The immune mechanisms that mediate protection against malaria are still unclear. Previously, we reported that mesenchymal stem cells (MSCs) play a critical role in host protection against malaria by altering the dynamic balance of T regulatory cells and effector T cells producing inflammatory cytokines. Here, we report that MSCs reprogram haematopoiesis in primary (bone marrow) and secondary (spleen) lymphoid organs to provide host protection against malaria. Adoptive transfer of MSCs from malaria-infected mice to naïve recipient mice that were subsequently infected with malaria parasites dramatically accelerated the formation of colony-forming units-erythroid cells in the bone marrow. Adoptively transferred MSCs also induced expression of the key erythroid cell differentiation factor GATA-1 in the spleen of recipient animals. Interestingly, we further observed a subtle increase in the CD34+ hematopoietic stem and progenitor cells in lymphoid organs, including spleen and lymph nodes. Infusion of MSCs also enhanced T cell proliferation, resulting in increased numbers of both CD4+ and CD8+ T cells in the spleen. MSCs also inhibited the induction of the negative co-stimulatory receptor programmed death-1 by T cells in recipient animals upon infection with malaria parasites. Taken together, our findings suggest that MSCs play a critical role in host protection against malaria infection by modulating erythropoiesis and lymphopoiesis.

7.
Mol Imaging ; 18: 1536012119852189, 2019.
Article in English | MEDLINE | ID: mdl-31187691

ABSTRACT

Expression of programmed cell death ligand 1 (PD-L1) within tumors is an important biomarker for guiding immune checkpoint therapies; however, immunohistochemistry-based methods of detection fail to provide a comprehensive picture of PD-L1 levels in an entire patient. To facilitate quantification of PD-L1 in the whole body, we developed a peptide-based, high-affinity PD-L1 imaging agent labeled with [18F]fluoride for positron emission tomography (PET) imaging. The parent peptide, WL12, and the nonradioactive analog of the radiotracer, 19FPy-WL12, inhibit PD-1/PD-L1 interaction at low nanomolar concentrations (half maximal inhibitory concentration [IC50], 26-32 nM). The radiotracer, [18F]FPy-WL12, was prepared by conjugating 2,3,5,6-tetrafluorophenyl 6-[18F]fluoronicotinate ([18F]FPy-TFP) to WL12 and assessed for specificity in vitro in 6 cancer cell lines with varying PD-L1 expression. The uptake of the radiotracer reflected the PD-L1 expression assessed by flow cytometry. Next, we performed the in vivo evaluation of [18F]FPy-WL12 in mice bearing cancer xenografts by PET imaging, ex vivo biodistribution, and blocking studies. In vivo data demonstrated a PD-L1-specific uptake of [18F]FPy-WL12 in tumors that is reduced in mice receiving a blocking dose. The majority of [18F]FPy-WL12 radioactivity was localized in the tumors, liver, and kidneys indicating the need for optimization of the labeling strategy to improve the in vivo pharmacokinetics of the radiotracer.


Subject(s)
B7-H1 Antigen/analysis , Fluorine Radioisotopes/chemistry , Peptides/chemistry , Positron-Emission Tomography/methods , Animals , Cell Line, Tumor , Humans , Radiochemistry
8.
ACS Appl Mater Interfaces ; 11(8): 7850-7861, 2019 Feb 27.
Article in English | MEDLINE | ID: mdl-30707559

ABSTRACT

Biomimetic nanoparticles (NPs) combine the flexibility and reproducibility of synthetic materials with the functionality of biological materials. Here, we developed and characterized biomimetic poly(lactic- co-glycolic acid) (PLGA) NPs coated with human cancer cell membrane fractions (CCMFs) to form CCMF-coated PLGA (CCMF-PLGA) NPs. We evaluated the ability of these CCMF-PLGA NPs to disrupt cancer cell-stromal cell interactions and to induce an immune response. Western blot analysis verified the plasma membrane purity of CCMFs. Confocal fluorescence microscopy and flow cytometry confirmed the presence of intact membrane-associated proteins including CXCR4 and CD44 following membrane derivation and coating. CCMFs and CCMF-PLGA NPs were capable of inhibiting cancer cell migration toward human mammary fibroblasts. Intravenous injection of CCMF-PLGA NPs significantly reduced experimental metastasis in vivo. Following immunization of Balb/c mice, near-infrared fluorescence imaging confirmed the migration of NPs to proximal draining lymph nodes (LNs). A higher percentage of CD8+ and CD4+ cytotoxic T-lymphocyte populations was observed in spleens and LNs of CCMF-PLGA NP-immunized mice. Splenocytes isolated from CCMF-PLGA NP-immunized mice had the highest number of interferon gamma-producing T-cells as detected by the ELISpot assay. CCMF-PLGA NPs hold promise for disrupting cancer cell-stromal cell interactions and for priming the immune system in cancer immunotherapy.


Subject(s)
Biomimetic Materials/therapeutic use , Cell Membrane/chemistry , Lung Neoplasms/prevention & control , Nanoparticles/chemistry , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Biomimetic Materials/pharmacology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Hyaluronan Receptors/chemistry , Interferon-gamma/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Receptors, CXCR4/chemistry
9.
J Clin Invest ; 129(2): 616-630, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30457978

ABSTRACT

Immune checkpoint therapies have shown tremendous promise in cancer therapy. However, tools to assess their target engagement, and hence the ability to predict their efficacy, have been lacking. Here, we show that target engagement and tumor-residence kinetics of antibody therapeutics targeting programmed death ligand-1 (PD-L1) can be quantified noninvasively. In computational docking studies, we observed that PD-L1-targeted monoclonal antibodies (atezolizumab, avelumab, and durvalumab) and a high-affinity PD-L1-binding peptide, WL12, have common interaction sites on PD-L1. Using the peptide radiotracer [64Cu]WL12 in vivo, we employed positron emission tomography (PET) imaging and biodistribution studies in multiple xenograft models and demonstrated that variable PD-L1 expression and its saturation by atezolizumab, avelumab, and durvalumab can be quantified independently of biophysical properties and pharmacokinetics of antibodies. Next, we used [64Cu]WL12 to evaluate the impact of time and dose on the unoccupied fraction of tumor PD-L1 during treatment. These quantitative measures enabled, by mathematical modeling, prediction of antibody doses needed to achieve therapeutically effective occupancy (defined as >90%). Thus, we show that peptide-based PET is a promising tool for optimizing dose and therapeutic regimens employing PD-L1 checkpoint antibodies, and can be used for improving therapeutic efficacy.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental , Peptides , Positron-Emission Tomography , Radiopharmaceuticals , A549 Cells , Animals , CHO Cells , Copper Radioisotopes , Cricetulus , Female , Humans , Male , Mice , Mice, Inbred NOD , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Peptides/chemistry , Peptides/pharmacokinetics , Peptides/pharmacology , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/pharmacology
10.
Mol Pharm ; 15(9): 3946-3952, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30037229

ABSTRACT

Tumors create and maintain an immunosuppressive microenvironment that promotes cancer cell escape from immune surveillance. The immune checkpoint protein programmed death-ligand 1 (PD-L1) is expressed in many cancers and is an important contributor to the maintenance of the immunosuppressive tumor microenvironment. PD-L1 is a prominent target for cancer immunotherapy. Guidance of anti-PD-L1 therapy is currently effected through measurement of PD-L1 through biopsy and immunohistochemistry. Here, we report a peptide-based imaging agent, [68Ga]WL12, to detect PD-L1 expression in tumors noninvasively by positron emission tomography (PET). WL12, a cyclic peptide comprising 14 amino acids, binds to PD-L1 with high affinity (IC50≈ 23 nM). Synthesis of [68Ga]WL12 provided radiochemical purity >99% after purification. Biodistribution in immunocompetent mice demonstrated 11.56 ± 3.18, 4.97 ± 0.8, 1.9 ± 0.1, and 1.33 ± 0.21 percentage of injected dose per gram (%ID/g) in hPD-L1, MDAMB231, SUM149, and CHO tumors, respectively, at 1 h postinjection, with high binding specificity noted with coinjection of excess, nonradiolabeled WL12. PET imaging demonstrated high tissue contrast in all tumor models tested.


Subject(s)
B7-H1 Antigen/metabolism , Gallium Radioisotopes/chemistry , Peptides/chemistry , Positron-Emission Tomography/methods , Triple Negative Breast Neoplasms/diagnostic imaging , Triple Negative Breast Neoplasms/metabolism , Animals , CHO Cells , Cricetulus , Female , Flow Cytometry , Immunohistochemistry , Mice
11.
Chemistry ; 24(28): 7235-7242, 2018 May 17.
Article in English | MEDLINE | ID: mdl-29508450

ABSTRACT

Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) is an innovative molecular imaging technique in which contrast agents are labeled by saturating their exchangeable proton spins by radio-frequency irradiation. Salicylic acid and its analogues are a promising class of highly sensitive, diamagnetic CEST agents. Herein, polymeric agents grafted with salicylic acid moieties and a known high-affinity ligand targeting prostate-specific membrane antigen in approximately 10:1 molar ratio were synthesized to provide sufficient MRI sensitivity and receptor specificity. The proton-exchange properties of the contrast agent in solution and in an experimental murine model are reported to demonstrate the feasibility of receptor-targeted CEST MRI of prostate cancer. Furthermore, the CEST imaging data were validated with an 111 In-labeled analogue of the agent by in vivo single photon emission computed tomographic imaging and tissue biodistribution studies.


Subject(s)
Contrast Media/chemistry , Polymers/chemistry , Prostatic Neoplasms/diagnostic imaging , Salicylic Acid/chemistry , Animals , Humans , Magnetic Resonance Imaging , Male , Protons , Tissue Distribution
12.
J Nucl Med ; 59(3): 486-493, 2018 03.
Article in English | MEDLINE | ID: mdl-29025989

ABSTRACT

Prostate-specific membrane antigen (PSMA) is highly expressed in prostate cancer and within the neovasculature of other solid tumors. The nonprostatic expression of PSMA has been reported exclusively within the neovasculature endothelial cells of nonprostatic cancers; however, there are few reports on PSMA expression in epithelial cells. Herein, we describe PSMA expression in nonprostatic epithelial cells and characterize the potential of PSMA-binding agents to noninvasively detect that expression. Methods: PSMA expression data were extracted from publicly available genomic databases. Genomic data were experimentally validated for PSMA expression-by quantitative reverse transcription polymerase chain reaction, flow cytometry, and Western blotting-in several nonprostatic cell lines and xenografts of melanoma and small cell lung cancer (SCLC) origin. The feasibility of PSMA detection in those tumor models was further established using PSMA-based nuclear and optical imaging agents and by biodistribution, blocking, and ex vivo molecular characterization studies. Results: We discovered that a small percentage of nonprostatic cancer cell lines and tumors express PSMA. Importantly, PSMA expression was sufficiently high to image established melanoma and SCLC xenografts using PSMA-based nuclear and optical imaging agents. Conclusion: These results indicate that PSMA expression in nonprostatic tumors may not be limited to the endothelium but may also include solid tumor tissue of nonprostatic cancers including melanoma and SCLC. Our observations indicate broader applicability of PSMA-targeted imaging and therapeutics.


Subject(s)
Gene Expression Regulation, Neoplastic , Glutamate Carboxypeptidase II/metabolism , Neoplasms/diagnostic imaging , Neoplasms/pathology , Single Photon Emission Computed Tomography Computed Tomography , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Humans , Male , Mice , Neoplasms/metabolism , Tissue Distribution
13.
Chemistry ; 23(58): 14469-14475, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-28771849

ABSTRACT

The CXCR4 chemokine receptor plays a key regulatory role in many biological functions, including embryonic development and controlling leukocyte functions during inflammation and immunity. CXCR4 has been also associated with multiple types of cancers where its overexpression/activation promotes metastasis, angiogenesis, and tumor growth and/or survival. Furthermore, CXCR4 is involved in HIV replication, as it is a co-receptor for viral entry into host cells. Altogether, these features make CXCR4 a very attractive target for the development of imaging and therapeutic agents. Here, the in vivo evaluation of the MCoTI-based cyclotide, MCo-CVX-5c, for the development of imaging agents that target CXCR4 is reported. Cyclotide MCo-CVX-5c is a potent CXCR4 antagonist with a remarkable in vivo resistance to biological degradation in serum. A [64 Cu]-DOTA-labeled version of this cyclotide demonstrated high and significant uptake in U87-stb-CXCR4 tumors compared to the control U87 tumors. Furthermore, protracted imaging studies demonstrated radiotracer retention in the U87-stb-CXCR4 tumor at 24 h post injection. Uptake in U87-stb-CXCR4 tumors could be blocked by unlabeled MCo-CVX-5c, showing high in vivo specificity. These results demonstrate the in vivo specificity and retention of a bioactive molecularly targeted cyclotide and highlight the potential of bioactive cyclotides for the development of new imaging agents that target CXCR4.


Subject(s)
Contrast Media/chemistry , Cyclotides/chemistry , Receptors, CXCR4/metabolism , Amino Acid Sequence , Animals , Brain Neoplasms/diagnosis , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Contrast Media/chemical synthesis , Contrast Media/metabolism , Cyclotides/chemical synthesis , Cyclotides/metabolism , Female , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Mice, SCID , Positron Emission Tomography Computed Tomography , Protein Binding , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Receptors, CXCR4/antagonists & inhibitors , Tissue Distribution , Transplantation, Heterologous
15.
Mol Imaging ; 16: 1536012117718459, 2017.
Article in English | MEDLINE | ID: mdl-28707500

ABSTRACT

Immunotherapy holds great promise in cancer treatment. The challenges in advancing immunotherapies lie in patient stratification and monitoring therapy. Noninvasive detection of immune checkpoint ligand PD-L1 can serve as an important biomarker for guidance and monitoring of immunotherapy. Here in, we provide an overview of our efforts to develop clinically translatable PD-L1-specific imaging agents for quantitative and real-time assessment of PD-L1 expression in tumor microenvironment.


Subject(s)
B7-H1 Antigen/metabolism , Immunotherapy/methods , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , B7-H1 Antigen/genetics , Biomarkers, Tumor/metabolism , Humans , Positron Emission Tomography Computed Tomography , Tomography, Emission-Computed, Single-Photon
16.
PLoS One ; 12(5): e0176958, 2017.
Article in English | MEDLINE | ID: mdl-28472168

ABSTRACT

Prostate specific membrane antigen (PSMA) targeted microbubbles (MBs) were developed using bioorthogonal chemistry. Streptavidin-labeled MBs were treated with a biotinylated tetrazine (MBTz) and targeted to PSMA expressing cells using trans-cyclooctene (TCO)-functionalized anti-PSMA antibodies (TCO-anti-PSMA). The extent of MB binding to PSMA positive cells for two different targeting strategies was determined using an in vitro flow chamber. The initial approach involved pretargeting, where TCO-anti-PSMA was first incubated with PSMA expressing cells and followed by MBTz, which subsequently showed a 2.8 fold increase in the number of bound MBs compared to experiments performed in the absence of TCO-anti-PSMA. Using direct targeting, where TCO-anti-PSMA was linked to MBTz prior to initiation of the assay, a 5-fold increase in binding compared to controls was observed. The direct targeting approach was subsequently evaluated in vivo using a human xenograft tumor model and two different PSMA-targeting antibodies. The US signal enhancements observed were 1.6- and 5.9-fold greater than that for non-targeted MBs. The lead construct was also evaluated in a head-to-head study using mice bearing both PSMA positive or negative tumors in separate limbs. The human PSMA expressing tumors exhibited a 2-fold higher US signal compared to those tumors deficient in human PSMA. The results demonstrate both the feasibility of preparing PSMA-targeted MBs and the benefits of using bioorthogonal chemistry to create targeted US probes.


Subject(s)
Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Microbubbles , Prostatic Neoplasms/immunology , Ultrasonics , Animals , Antibodies/immunology , Antigens, Surface/immunology , Glutamate Carboxypeptidase II/immunology , Heterografts , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/pathology
17.
Mod Pathol ; 30(4): 530-538, 2017 04.
Article in English | MEDLINE | ID: mdl-28059094

ABSTRACT

Predicting response to checkpoint blockade therapy for lung cancer has largely focused on measuring programmed death-ligand 1 (PD-L1) expression on tumor cells. PD-L1 expression is geographically heterogeneous within many tumors, however, and we questioned whether small tissue samples, such as biopsies, might be sufficiently representative of PD-L1 expression for evaluating this marker in lung cancer tumors. To evaluate the extent of variability of PD-L1 expression in small tissue samples, and how that variability affects accuracy of overall assessment of PD-L1 in lung cancer, we scored immunohistochemical staining for PD-L1 in tissue microarray cores from a series of 79 squamous cell lung cancers and 71 pulmonary adenocarcinomas. Our study found substantial inconsistencies for the percentages of cells staining positive for PD-L1 among different tissue microarray cores in many cases of both adenocarcinoma and squamous cell carcinoma. This variable scoring was seen at both high levels and low levels of PD-L1 expression, and by further evaluation of cases with discordant results on full-face sections to assess geographic distribution of staining, we found that discordant results among different tissue microarray cores reflected geographic variation of PD-L1 expression in those tumors. Moreover, we found that as a result of heterogeneous expression, the sensitivity of a single small tissue sample can be as low as 85% for detecting PD-L1 expression at scoring thresholds commonly used in clinical practice. Based on these studies, we conclude that many cases of lung cancer could be inaccurately or variably scored for PD-L1 expression with a single biopsy sample. Accordingly, lung cancer patients can be inconsistently classified for PD-L1 expression status, particularly when a threshold for the percentage of positive cells is used to determine eligibility for checkpoint blockade therapy.


Subject(s)
Adenocarcinoma/metabolism , B7-H1 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Squamous Cell/pathology , Female , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Male , Tissue Array Analysis
18.
J Photochem Photobiol B ; 167: 111-116, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28063300

ABSTRACT

Prostate-specific membrane antigen (PSMA) is over-expressed in the epithelium of prostate cancer and in the neovasculature of many non-prostate solid tumors. PSMA has been increasingly used as a target for cancer imaging and therapy. Here we describe a low-molecular-weight theranostic photosensitizer, YC-9, for PSMA-targeted optical imaging and photodynamic therapy (PDT). YC-9 was synthesized by conjugating IRDye700DX N-hydroxysuccinimide (NHS) ester with a PSMA targeting Lys-Glu urea through a lysine-suberate linker in suitable yield. Optical imaging in vivo demonstrated PSMA-specific tumor uptake of YC-9 with rapid clearance from non-target tissues. PSMA-specific cell kill was demonstrated with YC-9in vitro through PDT in PSMA+ PC3-PIP and PSMA- PC3-flu cells. In vivo PDT in mice bearing PSMA+ PC3-PIP tumors at 4h post-injection of YC-9 (A total of four PDT sessions were performed, 48h apart) resulted in significant tumor growth delay, while tumors in control groups continued to grow. PDT with YC-9 significantly increased the median survival of the PSMA+ PC3-PIP tumor mice (56.5days) compared to control groups [23.5-30.0days, including untreated, light alone, YC-9 alone (without light) and non-targeted IRDye700DX PDT treatment groups], without noticeable toxicity at the doses used. This study proves in principle that YC-9 is a promising therapeutic agent for targeted PDT of PSMA-expressing tissues, such as prostate tumors, and may also be useful against non-prostate tumors by virtue of neovascular PSMA expression.


Subject(s)
Photochemotherapy , Photosensitizing Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Theranostic Nanomedicine , Animals , Male , Mice , Mice, Inbred NOD , Mice, SCID , Optical Imaging , Photosensitizing Agents/chemical synthesis , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology
19.
Biochem Biophys Res Commun ; 483(1): 258-263, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28025143

ABSTRACT

Molecular imaging can report on the status of the tumor immune microenvironment and guide immunotherapeutic strategies to enhance the efficacy of immune modulation therapies. Imaging agents that can rapidly report on targets of immunomodulatory therapies are few. The programmed death ligand 1 (PD-L1) is an immune checkpoint protein over-expressed in several cancers and contributes to tumor immune suppression. Tumor PD-L1 expression is indicative of tumor response to PD-1 and PD-L1 targeted therapies. Herein, we report a highly specific peptide-based positron emission tomography (PET) imaging agent for PD-L1. We assessed the binding modes of the peptide WL12 to PD-L1 by docking studies, developed a copper-64 labeled WL12 ([64Cu]WL12), and performed its evaluation in vitro, and in vivo by PET imaging, biodistribution and blocking studies. Our results show that [64Cu]WL12 can be used to detect tumor PD-L1 expression specifically and soon after injection of the radiotracer, to fit within the standard clinical workflow of imaging within 60 min of administration.


Subject(s)
B7-H1 Antigen/analysis , Neoplasms/metabolism , Peptides/metabolism , Peptides/pharmacokinetics , Positron-Emission Tomography/methods , Animals , B7-H1 Antigen/metabolism , CHO Cells , Copper Radioisotopes/administration & dosage , Copper Radioisotopes/pharmacokinetics , Cricetulus , Diagnostic Uses of Chemicals , Female , Humans , Mice, SCID , Molecular Docking Simulation , Neoplasms/diagnostic imaging , Peptides/administration & dosage , Programmed Cell Death 1 Receptor/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
20.
Bioconjug Chem ; 27(9): 2103-10, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27458027

ABSTRACT

The programmed death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) pair is a major immune checkpoint pathway exploited by cancer cells to develop and maintain immune tolerance. With recent approvals of anti-PD-1 and anti-PD-L1 therapeutic antibodies, there is an urgent need for noninvasive detection methods to quantify dynamic PD-L1 expression in tumors and to evaluate the tumor response to immune modulation therapies. To address this need, we assessed [(64)Cu]atezolizumab for the detection of PD-L1 expression in tumors. Atezolizumab (MPDL3208A) is a humanized, human and mouse cross-reactive, therapeutic PD-L1 antibody that is being investigated in several cancers. Atezolizumab was conjugated with DOTAGA and radiolabeled with copper-64. The resulting [(64)Cu]atezolizumab was assessed for in vitro and in vivo specificity in multiple cell lines and tumors of variable PD-L1 expression. We performed PET-CT imaging, biodistribution, and blocking studies in NSG mice bearing tumors with constitutive PD-L1 expression (CHO-hPD-L1) and in controls (CHO). Specificity of [(64)Cu]atezolizumab was further confirmed in orthotopic tumor models of human breast cancer (MDAMB231 and SUM149) and in a syngeneic mouse mammary carcinoma model (4T1). We observed specific binding of [(64)Cu]atezolizumab to tumor cells in vitro, correlating with PD-L1 expression levels. Specific accumulation of [(64)Cu]atezolizumab was also observed in tumors with high PD-L1 expression (CHO-hPD-L1 and MDAMB231) compared to tumors with low PD-L1 expression (CHO, SUM149). Collectively, these studies demonstrate the feasibility of using [(64)Cu]atezolizumab for the detection of PD-L1 expression in different tumor types.


Subject(s)
Antibodies, Monoclonal , B7-H1 Antigen/metabolism , Copper Radioisotopes , Positron Emission Tomography Computed Tomography , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Biological Transport , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Female , Humans , Mice , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL