15 results on '"Raymond J. Moniz"'
Search Results
2. A versatile platform for generating engineered extracellular vesicles with defined therapeutic properties
- Author
-
Sriram Sathyanarayanan, Dalia Burzyn, Su Chul Jang, Sonya Haupt, Mcconnell Russell E, Scott Estes, Charan Leng, Ke Xu, James E. Thornton, Raymond J. Moniz, Kyriakos D. Economides, Nikki L. Ross, Youniss Madeleine, Rane A. Harrison, Olga Burenkova, Chang Ling Sia, Kevin Dooley, Martin Shelly, Jonathan D. Finn, Jorge Sanchez-Salazar, Kulman John D, Bryan D. Choi, Damian Houde, Christine McCoy, Nuruddeen D. Lewis, Katherine Kirwin, Leonid Gaidukov, and Williams Douglas E
- Subjects
Scaffold ,High density ,Nerve Tissue Proteins ,RNA-binding protein ,Cell Communication ,Extracellular vesicles ,Extracellular Vesicles ,Mice ,03 medical and health sciences ,Drug Delivery Systems ,0302 clinical medicine ,Drug Discovery ,Genetics ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Chemistry ,Cas9 ,Membrane Proteins ,Proteins ,Microvesicles ,Neoplasm Proteins ,Cell biology ,Repressor Proteins ,HEK293 Cells ,030220 oncology & carcinogenesis ,Commentary ,Molecular Medicine ,Female ,Intracellular ,Macromolecule - Abstract
Extracellular vesicles (EVs) are an important intercellular communication system facilitating the transfer of macromolecules between cells. Delivery of exogenous cargo tethered to the EV surface or packaged inside the lumen are key strategies for generating therapeutic EVs. We identified two "scaffold" proteins, PTGFRN and BASP1, that are preferentially sorted into EVs and enable high-density surface display and luminal loading of a wide range of molecules, including cytokines, antibody fragments, RNA binding proteins, vaccine antigens, Cas9, and members of the TNF superfamily. Molecules were loaded into EVs at high density and exhibited potent in vitro activity when fused to full-length or truncated forms of PTGFRN or BASP1. Furthermore, these engineered EVs retained pharmacodynamic activity in a variety of animal models. This engineering platform provides a simple approach to functionalize EVs with topologically diverse macromolecules and represents a significant advance toward unlocking the therapeutic potential of EVs.
- Published
- 2021
- Full Text
- View/download PDF
3. ExoSTING, an extracellular vesicle loaded with STING agonists, promotes tumor immune surveillance
- Author
-
Kevin Dooley, Nuruddeen D. Lewis, Raymond W. Bourdeau, Williams Douglas E, Christine McCoy, Kelvin Xi Zhang, Katherine Kirwin, Joyoti Dey, Marc Grenley, Jorge Sanchez-Salazar, Nikki L. Ross, Agata Villiger-Oberbek, Su Chul Jang, Sriram Sathyanarayanan, Tong Zi, Joanne Lim, Kyriakos D. Economides, Ke Xu, William K. Dahlberg, Raymond J. Moniz, Scott Estes, Rane A. Harrison, and Chang Ling Sia
- Subjects
0301 basic medicine ,QH301-705.5 ,Medicine (miscellaneous) ,Cancer immunotherapy ,Systemic inflammation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,Extracellular Vesicles ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Tumor Microenvironment ,medicine ,Animals ,Biology (General) ,Antigen-presenting cell ,Immunologic Surveillance ,Mice, Inbred BALB C ,Tumor microenvironment ,Extracellular vesicle ,Extravasation ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,Stimulator of interferon genes ,Cancer research ,Nanoparticles ,Female ,medicine.symptom ,General Agricultural and Biological Sciences - Abstract
Cyclic dinucleotide (CDN) agonists of the STimulator of InterferoN Genes (STING) pathway have shown immune activation and tumor clearance in pre-clinical models. However, CDNs administered intratumorally also promote STING activation leading to direct cytotoxicity of many cell types in the tumor microenvironment (TME), systemic inflammation due to rapid tumor extravasation of the CDN, and immune ablation in the TME. These result in a failure to establish immunological memory. ExoSTING, an engineered extracellular vesicle (EV) exogenously loaded with CDN, enhances the potency of CDN and preferentially activates antigen presenting cells in the TME. Following intratumoral injection, exoSTING was retained within the tumor, enhanced local Th1 responses and recruitment of CD8+ T cells, and generated systemic anti-tumor immunity to the tumor. ExoSTING at therapeutically active doses did not induce systemic inflammatory cytokines, resulting in an enhanced therapeutic window. ExoSTING is a novel, differentiated therapeutic candidate that leverages the natural biology of EVs to enhance the activity of CDNs., Su Chul Jang et al. develop exoSTING, consisting of an engineered extracellular vesicle loaded with a potent cyclic dinucleotide (CDN) agonist of the STING pathway. They find that exoSTING shows more than 100-fold increased potency in in vivo tumor models and has increased tumor retention and lower levels of systemic inflammatory cytokine production as compared to free CDN.
- Published
- 2021
- Full Text
- View/download PDF
4. Characterization of murine CEACAM1 in vivo reveals low expression on CD8+ T cells and no tumor growth modulating activity by anti-CEACAM1 mAb CC1
- Author
-
Liming Liu, Jason Laskey, Raymond J. Moniz, Toya Nath Baral, Joann A. O'Connor, Archie Tse, Andrey Loboda, Robbie L. McLeod, Mike Lee, Minilik Angagaw, Jin-Hwan Han, SuChun Hsieh, Juha Punnonen, Sarah Javaid, Svetlana Sadekova, and Hassan Issafras
- Subjects
0301 basic medicine ,Chemistry ,medicine.drug_class ,Cell adhesion molecule ,medicine.medical_treatment ,CC1 ,CD8+ T cells ,Monoclonal antibody ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,Cancer immunotherapy ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Cytotoxic T cell ,syngeneic mouse model ,pharmacokinetic ,Signal transduction ,CEACAM1 ,CD8 ,Research Paper - Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) has been reported to mediate both tumorigenic and anti-tumor effects in vivo. Blockade of the CEACAM1 signaling pathway has recently been implicated as a novel mechanism for cancer immunotherapy. CC1, a mouse anti-CEACAM1 monoclonal antibody (mAb), has been widely used as a pharmacological tool in preclinical studies to inform on CEACAM1 pathway biology although limited data are available on its CEACAM1 blocking characteristics or pharmacodynamic-pharmacokinetic profiles. We sought to investigate CEACAM1 expression on mouse tumor and immune cells, characterize CC1 mAb binding, and evaluate CC1 in syngeneic mouse oncology models as a monotherapy and in combination with an anti-PD-1 mAb. CEACAM1 expression was observed at high levels on neutrophils, NK cells and myeloid-derived suppressor cells (MDSCs), while the expression on tumor-infiltrating CD8+ T cells was low. Unexpectedly, rather than blocking, CC1 facilitated binding of soluble CEACAM1 to CEACAM1 expressing cells. No anti-tumor effects were observed in CT26, MBT2 or A20 models when tested up to 30 mg/kg dose, a dose that was estimated to achieve >90% target engagement in vivo. Taken together, tumor infiltrating CD8+ T cells express low levels of CEACAM1 and CC1 Ab mediates no or minimal anti-tumor effects in vivo, as a monotherapy or in combination with anti-PD-1 treatment.
- Published
- 2018
- Full Text
- View/download PDF
5. A vault nanoparticle vaccine induces protective mucosal immunity.
- Author
-
Cheryl I Champion, Valerie A Kickhoefer, Guangchao Liu, Raymond J Moniz, Amanda S Freed, Liisa L Bergmann, Dana Vaccari, Sujna Raval-Fernandes, Ann M Chan, Leonard H Rome, and Kathleen A Kelly
- Subjects
Medicine ,Science - Abstract
Generation of robust cell-mediated immune responses at mucosal surfaces while reducing overall inflammation is a primary goal for vaccination. Here we report the use of a recombinant nanoparticle as a vaccine delivery platform against mucosal infections requiring T cell-mediated immunity for eradication.We encapsulated an immunogenic protein, the major outer membrane protein (MOMP) of Chlamydia muridarum, within hollow, vault nanocapsules (MOMP-vaults) that were engineered to bind IgG for enhanced immunity. Intranasal immunization (i.n) with MOMP-vaults induced anti-chlamydial immunity plus significantly attenuated bacterial burden following challenge infection. Vault immunization induced anti-chlamydial immune responses and inflammasome formation but did not activate toll-like receptors. Moreover, MOMP-vault immunization enhanced microbial eradication without the inflammation usually associated with adjuvants.Vault nanoparticles containing immunogenic proteins delivered to the respiratory tract by the i.n. route can act as "smart adjuvants" for inducing protective immunity at distant mucosal surfaces while avoiding destructive inflammation.
- Published
- 2009
- Full Text
- View/download PDF
6. Effect of cryopreservation on delineation of immune cell subpopulations in tumor specimens as determined by multiparametric single cell mass cytometry analysis
- Author
-
Ilona Kariv, Raymond J. Moniz, Ying Huo, Elma Kadic, and An Chi
- Subjects
0301 basic medicine ,Carcinogenesis ,Immunology ,Cell Separation ,CyTOF™ ,Biology ,Mass Spectrometry ,Immunophenotyping ,Flow cytometry ,03 medical and health sciences ,Mass Cytometry (MC) ,Tumor Microenvironment ,medicine ,Humans ,Cell Lineage ,Mass cytometry ,Tumor microenvironment (TME) ,Carcinoma, Renal Cell ,Cryopreservation ,Tumor microenvironment ,medicine.diagnostic_test ,Tumor samples ,Myeloid-Derived Suppressor Cells ,Lineage markers ,Cell Differentiation ,Molecular biology ,Phenotype ,030104 developmental biology ,Tumor progression ,ViSNE ,Myeloid-derived Suppressor Cell ,Feasibility Studies ,Tumor infiltrating lymphocytes (TILs) ,Single-Cell Analysis ,Colorectal Neoplasms ,Cytometry ,Research Article ,Flow cytometry (FC) - Abstract
Background Comprehensive understanding of cellular immune subsets involved in regulation of tumor progression is central to the development of cancer immunotherapies. Single cell immunophenotyping has historically been accomplished by flow cytometry (FC) analysis, enabling the analysis of up to 18 markers. Recent advancements in mass cytometry (MC) have facilitated detection of over 50 markers, utilizing high resolving power of mass spectrometry (MS). This study examined an analytical and operational feasibility of MC for an in-depth immunophenotyping analysis of the tumor microenvironment, using the commercial CyTOF™ instrument, and further interrogated challenges in managing the integrity of tumor specimens. Results Initial longitudinal studies with frozen peripheral blood mononuclear cells (PBMCs) showed minimal MC inter-assay variability over nine independent runs. In addition, detection of common leukocyte lineage markers using MC and FC detection confirmed that these methodologies are comparable in cell subset identification. An advanced multiparametric MC analysis of 39 total markers enabled a comprehensive evaluation of cell surface marker expression in fresh and cryopreserved tumor samples. This comparative analysis revealed significant reduction of expression levels of multiple markers upon cryopreservation. Most notably myeloid derived suppressor cells (MDSC), defined by co-expression of CD66b+ and CD15+, HLA-DRdim and CD14− phenotype, were undetectable in frozen samples. Conclusion These results suggest that optimization and evaluation of cryopreservation protocols is necessary for accurate biomarker discovery in frozen tumor specimens. Electronic supplementary material The online version of this article (doi:10.1186/s12865-017-0192-1) contains supplementary material, which is available to authorized users.
- Published
- 2017
- Full Text
- View/download PDF
7. Abstract 944: exoSTING: An engineered exosome therapeutic that selectively delivers STING agonist to the tumor resident antigen-presenting cells resulting in improved tumor antigen-specific adaptive immune response
- Author
-
Kevin Dooley, Rane A. Harrison, Christine McCoy, Chang Ling Sia, Raymond J. Moniz, Damian Houde, Tong Zi, Scott Estes, Ke Xu, Nikki L. Ross, Raymond W. Bourdeau, Kyriakos D. Economides, Sriram Sathyanarayanan, Agata Villiger-Oberbek, Nuruddeen D. Lewis, Jorge Sanchez-Salazar, William K. Dahlberg, and Su Chul Jang
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,medicine.medical_treatment ,Acquired immune system ,Exosome ,eye diseases ,Microvesicles ,Tumor antigen ,03 medical and health sciences ,Sting ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,Immune system ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Antigen-presenting cell ,business - Abstract
Background: The Stimulator of Interferon Genes (STING) pathway is an attractive target in immuno-oncology. Selective activation of the STING pathway in antigen presenting cells (APCs) is essential for eliciting a potent and specific anti-tumor immune response. STING is ubiquitously expressed in normal cells including T cells and endothelial cells. Direct intra-tumoral administration of a free STING agonist results in activation of the STING pathway in all cells, resulting in loss of viability of immune cells, tissue damage, and systemic immune activation due to vascular leakage. Exosomes are an efficient natural messenger system that delivers macromolecules between cells. Leveraging exosome biology, we have developed a novel engineered exosome therapeutic, exoSTING, to selectively target the STING pathway in tumor resident APCs. Results: exoSTING is composed of exosomes engineered to express high levels of Protein X (PrX), a transmembrane glycoprotein naturally occurring on exosomes, and loaded with a STING agonist. Following intra-tumoral injection in checkpoint refractory B16F10 tumors, exoSTING selectively activates STING in APCs, leading to anti-tumor immunity with greater than 100-fold improvement in potency compared to free STING agonist. The activity of exoSTING is substantially diminished in the PrX knock out exosomes, highlighting the importance of this surface glycoprotein for preferential activation of APCs. In contrast to the free STING agonist, exoSTING is retained within the injected tumor, minimizing systemic exposure. Furthermore, exoSTING administration preserves the viability of T cells and APCs, reduces collateral tissue damage, and does not induce systemic cytokine production, resulting in a broader therapeutic window in contrast to free STING agonist. exoSTING produced an increased systemic tumor-specific T cell response as demonstrated by elimination of non-injected abscopal tumors. The specificity of exoSTING activity was demonstrated using a STING knockout mouse (Tmem173gt/J). T cells and myeloid cells play a critical role in exoSTING mediated anti-tumor immunity, in contrast to NK cells which are not required. exoSTING treatment results in significant induction of PD-L1 expression (P Conclusion: exoSTING is an engineered exosome therapeutic that leverages exosome biology and specifically targets the STING pathway in APCs, resulting in greater potency with preserved viability of T cells and APCs, greater systemic tumor antigen specific immune response, reduced systemic cytokine production, and enhanced efficacy. Citation Format: Su Chul Jang, Raymond J. Moniz, Chang Ling Sia, Rane A. Harrison, Damian Houde, Nikki Ross, Ke Xu, Nuruddeen Lewis, Raymond Bourdeau, Christine McCoy, Tong Zi, Agata Villiger-Oberbek, Scott Estes, Jorge Sanchez-Salazar, Kevin Dooley, William K. Dahlberg, Sriram Sathyanarayanan, Kyriakos D. Economides. exoSTING: An engineered exosome therapeutic that selectively delivers STING agonist to the tumor resident antigen-presenting cells resulting in improved tumor antigen-specific adaptive immune response [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 944.
- Published
- 2019
- Full Text
- View/download PDF
8. Abstract LB-012: Single cell RNA-sequencing of multiple myeloma clinical patients for identifying novel immuno-oncology drug targets
- Author
-
Raymond J. Moniz, Namit Kumar, Kelvin Xi Zhang, Lixia Li, Vanessa M. Peterson, Philip W. Garrett-engele, and Joel A. Klappenbach
- Subjects
Melphalan ,Cancer Research ,biology ,CD52 ,business.industry ,Cell ,Cancer ,medicine.disease ,Biomarker (cell) ,Transcriptome ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Medicine ,Antibody ,business ,Multiple myeloma ,medicine.drug - Abstract
Recent advances in single cell genomic technologies have enabled unbiased quantification of cellular states within complex tissues, mechanisms of differentiation, biomarker presentation in cellular subsets; yet their applications in evaluating the response to therapeutic interventions are still underexplored. In our study, we apply single cell RNA sequencing (scRNA-seq) to uncover mechanisms and drug targets for Multiple Myeloma(MM), a malignancy of plasma cells with estimates of 30,770 new cases and 12,770 deaths in 2018 within US alone. Using the 10x Genomics scRNA-seq platform, we profiled ten clinical MM patients, pre- and post-treatment with four standard of care chemotherapy regimens (VAD, VCD, VMP, MP; A=Adriamycin; C=Cyclophosphamide; D=Dexamethasone; M=Melphalan; P=Prednisone; V=Velcade). We sequenced a total of 106,000 cells and determined differential gene expression upon treatment at single-cell resolution comparing 37,926 pre- vs 44,842 post-treatment cells. We identify activation of immune cells with increased expression of IL1B, CD14, CD52, CCL4, IFITM3, PSMA4; and repression of translational machinery with reduced expression of PABPC1, PABPC4, FUS, POLR2A in post-treatment (abs(log2 FC >0.25) and P-value < 10-5). To understand the functional implication of gene expression changes, we identified that the upregulated genes are enriched for ontologies regulating protein ubiquitination pathway, communication between innate and adaptive immune cells, TREM1 signaling, dendritic cell maturation (P-value 3.16x10-17, 3.47x10-7, 3.09 x10-5, 4.07x10-5 respectively). Downregulated genes are enriched for EIF2 signaling, regulation of eIF4 and p70S6K signaling, mTOR signaling (P-value 3.16x10-13, 1.86x10-8, 2.24x10-6 respectively). Additionally, we performed a comparative analysis of differentially expressed genes in response to each drug regimen. We identified genes upregulated upon MP drug regimen are comparatively enriched for IL-8 signaling, while treatment with VMP enhances B-cell receptor signaling (P-value 5.5x10-12 and 9.0x10-9 respectively). Further, we have used our unbiased transcriptomic interrogation of MM clinical patients to identify a panel of 100+ biomarkers that are being evaluated at a proteomics level using our recently developed technique, RNA Expression and Protein sequencing (REAP-seq). This method enables us to obtain readout for whole transcriptome and proteins (>100) using oligo conjugated antibodies while retaining single cell resolution. Transcriptome and/or proteome analysis of bulk tumor often fails to detect signature of rare cell populations. Our study here highlights the use of single cell readouts for identifying novel oncology mechanisms, drug targets and drug combinations for more efficacious immunotherapies. Citation Format: Namit Kumar, Vanessa M. Peterson, Kelvin X. Zhang, Lixia Li, Philip W. Garrett-Engele, Raymond J. Moniz, Joel A. Klappenbach. Single cell RNA-sequencing of multiple myeloma clinical patients for identifying novel immuno-oncology drug targets [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr LB-012.
- Published
- 2018
- Full Text
- View/download PDF
9. Plasmacytoid dendritic cells modulate nonprotective T-cell responses to genital infection byChlamydia muridarum
- Author
-
Moshe Arditi, Raymond J. Moniz, Lynn K. Gordon, Kathleen A. Kelly, Jonathan Braun, and Ann M. Chan
- Subjects
Microbiology (medical) ,Chlamydia muridarum ,T-Lymphocytes ,T cell ,Immunology ,macromolecular substances ,T-Lymphocytes, Regulatory ,Microbiology ,Article ,Mice ,Immune system ,medicine ,Animals ,Humans ,Immunology and Allergy ,Mice, Inbred BALB C ,Chlamydia ,biology ,FOXP3 ,Forkhead Transcription Factors ,hemic and immune systems ,Dendritic Cells ,General Medicine ,Dendritic cell ,Chlamydia Infections ,Th1 Cells ,Natural killer T cell ,biology.organism_classification ,medicine.disease ,Infectious Diseases ,medicine.anatomical_structure ,Female ,Genital Diseases, Female ,CD8 - Abstract
Given their immune-modulating capacity, regulatory T cells (Treg) cells may be important players in the induction of the protective T-cell response (Th1) to genital chlamydial infection. Recent work has demonstrated that plasmacytoid dendritic cells (pDC) respond to genital chlamydial infection, and that pDC may be uniquely positioned for the induction of Treg cells during this infection. Here, we present the first data demonstrating that Treg influx into the draining lymph node and the site of infection during genital chlamydial infection. We found that pDC depletion altered the numbers of Treg and nonprotective inflammatory cells [interferongamma-(IFNgamma)-producing CD8+ T and IFNgamma-producing natural killer T cells] in the spleens of mice genitally infected with Chlamydia muridarum. Furthermore, pDC depletion did not alter Th1 cell numbers, indicating that pDC modulate cells that could inhibit and promote nonprotective inflammation during genital chlamydial infection. Finally, we demonstrate that depletion of pDC results in less severe dilation and collagen deposition in the oviduct following resolution of infection, implicating pDC activity in the formation of sequelae following genital C. muridarum infection.
- Published
- 2010
- Full Text
- View/download PDF
10. Blockade of epithelial membrane protein 2 (EMP2) abrogates infection ofChlamydia muridarummurine genital infection model
- Author
-
Catherine P. Coulam, Raymond J. Moniz, Kathleen A. Kelly, Eric M. Lepin, Ann M. Chan, Agnes K. Nagy, Anna M. Wu, Lynn K. Gordon, Madhuri Wadehra, Sergey Mareninov, Jonathan Braun, and Kaori Shimazaki
- Subjects
Microbiology (medical) ,Chlamydia muridarum ,Immunology ,Colony Count, Microbial ,Immunoglobulins ,medicine.disease_cause ,Microbiology ,Article ,Proinflammatory cytokine ,Mice ,In vivo ,medicine ,Animals ,Immunology and Allergy ,Chlamydiaceae ,Inflammation ,Infectivity ,Mice, Inbred BALB C ,Membrane Glycoproteins ,Chlamydia ,biology ,General Medicine ,Chlamydia Infections ,medicine.disease ,biology.organism_classification ,Infectious Diseases ,biology.protein ,Cytokines ,Female ,Antibody ,Chlamydia trachomatis ,Genital Diseases, Female - Abstract
New methods are needed to eradicate or prevent Chlamydia trachomatis infections. Blockade of epithelial membrane protein 2 (EMP2) by genetic silencing or neutralizing polyclonal antibody reduced chlamydial infectivity in vitro. This study tests the prediction that recombinant anti-EMP2 diabody could reduce early chlamydial infection of the genital tract in vivo. In a murine infection model, pretreatment with anti-EMP2 diabody, as compared with control diabody, significantly reduced bacterial load, tissue production of inflammatory cytokines, recruitment of polymorphonuclear leukocytes, and local tissue inflammation. These findings support EMP2 as a potential preventative and therapeutic target for genital chlamydial infection.
- Published
- 2009
- Full Text
- View/download PDF
11. Human anti-CAIX antibodies mediate immune cell inhibition of renal cell carcinoma in vitro and in a humanized mouse model in vivo
- Author
-
Zhongyao Xu, De-Kuan Chang, Quan Zhu, Raymond J. Moniz, Sabina Signoretti, Jiusong Sun, and Wayne A. Marasco
- Subjects
Cancer Research ,Carbonic anhydrase IX ,medicine.drug_class ,medicine.medical_treatment ,NSG mouse ,Antibody engineering ,Biology ,Monoclonal antibody ,Protein Engineering ,Mice ,Antigen ,In vivo ,Antigens, Neoplasm ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Lymphocytes ,Carcinoma, Renal Cell ,Carbonic Anhydrases ,Cell Proliferation ,Antibody-dependent cell-mediated cytotoxicity ,Cell growth ,Research ,Xenograft ,Antibody-Dependent Cell Cytotoxicity ,Antibodies, Monoclonal ,Immunotherapy ,Endocytosis ,Kidney Neoplasms ,Killer Cells, Natural ,Disease Models, Animal ,Oncology ,Immunology ,Humanized mouse ,Cancer research ,Molecular Medicine ,Orthotopic tumor ,Single-Chain Antibodies - Abstract
Background Carbonic anhydrase (CA) IX is a surface-expressed protein that is upregulated by the hypoxia inducible factor (HIF) and represents a prototypic tumor-associated antigen that is overexpressed on renal cell carcinoma (RCC). Therapeutic approaches targeting CAIX have focused on the development of CAIX inhibitors and specific immunotherapies including monoclonal antibodies (mAbs). However, current in vivo mouse models used to characterize the anti-tumor properties of fully human anti-CAIX mAbs have significant limitations since the role of human effector cells in tumor cell killing in vivo is not directly evaluated. Methods The role of human anti-CAIX mAbs on CAIX+ RCC tumor cell killing by immunocytes or complement was tested in vitro by antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and antibody-dependent cellular phagocytosis (ADCP) as well as on CAIX+ RCC cellular motility, wound healing, migration and proliferation. The in vivo therapeutic activity mediated by anti-CAIX mAbs was determined by using a novel orthotopic RCC xenograft humanized animal model and analyzed by histology and FACS staining. Results Our studies demonstrate the capacity of human anti-CAIX mAbs that inhibit CA enzymatic activity to result in immune-mediated killing of RCC, including nature killer (NK) cell-mediated ADCC, CDC, and macrophage-mediated ADCP. The killing activity correlated positively with the level of CAIX expression on RCC tumor cell lines. In addition, Fc engineering of anti-CAIX mAbs was shown to enhance the ADCC activity against RCC. We also demonstrate that these anti-CAIX mAbs inhibit migration of RCC cells in vitro. Finally, through the implementation of a novel orthotopic RCC model utilizing allogeneic human peripheral blood mononuclear cells in NOD/SCID/IL2Rγ−/− mice, we show that anti-CAIX mAbs are capable of mediating human immune response in vivo including tumor infiltration of NK cells and activation of T cells, resulting in inhibition of CAIX+ tumor growth. Conclusions Our findings demonstrate that these novel human anti-CAIX mAbs have therapeutic potential in the unmet medical need of targeted killing of HIF-driven CAIX+RCC. The orthotopic tumor xenografted humanized mouse provides an improved model to evaluate the in vivo anti-tumor capabilities of fully human mAbs for RCC therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0384-3) contains supplementary material, which is available to authorized users.
- Published
- 2015
12. Targeting vault nanoparticles to specific cell surface receptors
- Author
-
Muri Han, Mariena Silvestry, Kathleen A. Kelly, Sujna Raval-Fernandes, Leonard H. Rome, Valerie A. Kickhoefer, Dana Vaccari, Raymond J. Moniz, Michael J. Poderycki, and Phoebe L. Stewart
- Subjects
General Physics and Astronomy ,Peptide ,Receptors, Cell Surface ,Epitope ,Article ,Cell membrane ,Epitopes ,Mice ,Protein structure ,Epidermal growth factor ,Major vault protein ,Cell Line, Tumor ,medicine ,Animals ,Humans ,General Materials Science ,Vault (organelle) ,chemistry.chemical_classification ,Binding Sites ,biology ,Cell Membrane ,General Engineering ,Dendritic Cells ,Molecular biology ,Amino acid ,Cell biology ,Protein Structure, Tertiary ,ErbB Receptors ,medicine.anatomical_structure ,chemistry ,Immunoglobulin G ,biology.protein ,Nanoparticles ,HeLa Cells ,Protein Binding - Abstract
As a naturally occurring nanocapsule abundantly expressed in nearly all-eukaryotic cells, the barrel-shaped vault particle is perhaps an ideal structure to engineer for targeting to specific cell types. Recombinant vault particles self-assemble from 96 copies of the major vault protein (MVP), have dimensions of 72.5 × 41 nm, and have a hollow interior large enough to encapsulate hundreds of proteins. In this study, three different tags were engineered onto the C-terminus of MVP: an 11 amino acid epitope tag, a 33 amino acid IgG-binding peptide, and the 55 amino acid epidermal growth factor (EGF). These modified vaults were produced using a baculovirus expression system. Our studies demonstrate that recombinant vaults assembled from MVPs containing C-terminal peptide extensions, display these tags at the top and bottom of the vault on the outside of the particle, and can be used to specifically bind the modified vaults to epithelial cancer cells (A431) via the epidermal growth factor receptor (EGFR), either directly (EGF modified vaults) or as mediated by a monoclonal antibody (anti-EGFR) bound to recombinant vaults containing the IgG-binding peptide. The ability to target vaults to specific cells represents an essential advance towards using recombinant vaults as delivery vehicles.
- Published
- 2009
13. Identification of dendritic cell subsets responding to genital infection by Chlamydia muridarum
- Author
-
Kathleen A. Kelly, Ann M. Chan, and Raymond J. Moniz
- Subjects
Microbiology (medical) ,Chlamydia muridarum ,Immunology ,Plasmacytoid dendritic cell ,macromolecular substances ,Biology ,Microbiology ,Article ,Mice ,In vivo ,medicine ,Immunology and Allergy ,Animals ,Lymph node ,Mice, Inbred BALB C ,Chlamydia ,hemic and immune systems ,General Medicine ,Dendritic cell ,T lymphocyte ,Dendritic Cells ,Chlamydia Infections ,biology.organism_classification ,medicine.disease ,Infectious Diseases ,medicine.anatomical_structure ,Cytokines ,Female ,Genital Diseases, Female ,Conventional Dendritic Cell - Abstract
Dendritic cells (DCs) are central for the induction of T-cell responses needed for chlamydial eradication. Here, we report the activation of two DC subsets: a classical CD11b+ (cDC) and plasmacytoid (pDC) during genital infection with Chlamydia muridarum. Genital infection induced an influx of cDC and pDC into the genital tract and its draining lymph node (iliac lymph nodes, ILN) as well as colocalization with T cells in the ILN. Genital infection with C. muridarum also stimulated high levels of costimulatory molecules on cDC central for the activation of naive T cells in vivo. In contrast, pDC expressed low levels of most costimulatory molecules in vivo and did not secrete cytokines associated with the production of T helper (Th)1 cells in vitro. However, pDC upregulated inducible costimulatory ligand expression and produced IL-6 and IL-10 in response to chlamydial exposure in vitro. Our findings show that these two DC subsets likely have different functions in vivo. cDCs are prepared for induction of antichlamydial T-cell responses, whereas pDCs have characteristics associated with the differentiation of non-Th1 cell subsets.
- Published
- 2009
14. Abstract 269: Evaluation of the antitumor activity of anti-PD-1 immunotherapy as a single agent and in combination with approved agents in preclinical tumor models
- Author
-
Elaine M. Pinheiro, Ruban Mangadu, Brian Long, Mingmei Cai, Uyen Truong Phan, Heather Hirsch, Samik Basu, Ali-Samer Al-Assaad, Joseph H. Phillips, Yanhong Ma, Yaolin Wang, Venkataraman Sriram, Raymond J. Moniz, and Terrill K. McClanahan
- Subjects
Cancer Research ,biology ,business.industry ,medicine.medical_treatment ,Pembrolizumab ,Immunotherapy ,Pharmacology ,Blockade ,Clinical trial ,Immune system ,Oncology ,Pharmacodynamics ,Monoclonal ,biology.protein ,Cancer research ,Medicine ,Antibody ,business - Abstract
Pembrolizumab (MK-3475), a humanized monoclonal IgG4 antibody against programmed death receptor 1 (PD-1), is currently being studied in clinical trials across more than 30 types of cancers. To further support the clinical development of pembrolizumab and to aid the mechanistic understanding of anti-PD-1 immunotherapy, we generated a surrogate PD-1-blocking antibody (muDX400). We have used muDX400 to determine the antitumor activity, pharmacokinetics, and pharmacodynamics of PD-1 inhibition in multiple preclinical syngeneic tumor model systems. Response to muDX400 treatment in several syngeneic tumor models was broadly classified into 3 categories: highly responsive (ie, complete and durable tumor regressions were observed), partially responsive (ie, tumor growth inhibition was observed), and intrinsically resistant to therapy. Gene and protein expression signatures revealed that the more responsive models expressed higher levels of both PD-1 ligand (PD-L1) and tumor-infiltrating lymphocytes compared with nonresponsive models. To further evaluate mechanisms that could potentially enhance the antitumor activity of anti-PD-1 in these tumor models, muDX400 was combined with a number of different chemotherapies, targeted therapies, and other immunotherapies. Because immune suppression is a common side effect associated with many standard-of-care therapies, we evaluated the potential abrogation of muDX400-mediated antitumor activity when combined with approved therapies by scheduling the dosing regimen to examine concurrent and sequential administration of these agents. In the models in which enhanced antitumor activity was evident, we evaluated the immune landscape of blood, tumors, and draining lymph nodes by immuno-phenotyping and molecular profiling. These data provide preclinical support to expand the clinical development of pembrolizumab into additional cancer types as both a single agent and in combination with other approved anticancer therapies. Additional studies with muDX400 are ongoing to further elucidate the mechanism of action of PD-1 blockade and to better understand the antitumor responses observed in clinical trials of pembrolizumab. Citation Format: Elaine M. Pinheiro, Ruban Mangadu, Uyen T. Phan, Mingmei Cai, Yanhong Ma, Heather A. Hirsch, Terrill K. McClanahan, Raymond J. Moniz, Ali-Samer Al-Assaad, Samik Basu, Yaolin Wang, Venkataraman Sriram, Joseph H. Phillips, Brian J. Long. Evaluation of the antitumor activity of anti-PD-1 immunotherapy as a single agent and in combination with approved agents in preclinical tumor models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 269. doi:10.1158/1538-7445.AM2015-269
- Published
- 2015
- Full Text
- View/download PDF
15. Su.25. Mucosal Host Receptor Diabody Therapy for Chlamydia Infection in the Murine Genital Tract
- Author
-
Ann Chan, Anna M. Wu, Eric J. Lepin, Kaori Shimazaki, Sergey Mareninov, Jonathan Braun, Paige Coulam, Kathleen A. Kelly, James D. Marks, Raymond J. Moniz, Agnes K. Nagy, and Lynn K. Gordon
- Subjects
Chlamydia ,business.industry ,Host (biology) ,Genital tract ,Immunology ,Immunology and Allergy ,Medicine ,business ,medicine.disease ,Receptor ,Virology - Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.