40 results on '"Benjamin J. Capoccia"'
Search Results
2. Figure S2 from Development of AO-176, a Next-Generation Humanized Anti-CD47 Antibody with Novel Anticancer Properties and Negligible Red Blood Cell Binding
- Author
-
Daniel S. Pereira, Robert W. Karr, William A. Frazier, Pamela T. Manning, Michael J. Donio, Benjamin J. Capoccia, Ronald R. Hiebsch, Myriam N. Bouchlaka, and Robyn J. Puro
- Abstract
Figure S2 shows that AO-176 does not bind to mouse CD47
- Published
- 2023
- Full Text
- View/download PDF
3. Development of AO-176, a Next-Generation Humanized Anti-CD47 Antibody with Novel Anticancer Properties and Negligible Red Blood Cell Binding
- Author
-
Pamela T. Manning, Ronald R. Hiebsch, Daniel S. Pereira, Benjamin J. Capoccia, Robyn J Puro, Michael J. Donio, Myriam N. Bouchlaka, William A. Frazier, and Karr Robert W
- Subjects
0301 basic medicine ,Cancer Research ,Erythrocytes ,T-Lymphocytes ,Mice, Nude ,Apoptosis ,CD47 Antigen ,Antibodies, Monoclonal, Humanized ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Phagocytosis ,Antigen ,Neoplasms ,Tumor Cells, Cultured ,Signal-regulatory protein alpha ,Animals ,Humans ,Receptors, Immunologic ,Cell Proliferation ,Antibody-dependent cell-mediated cytotoxicity ,Mice, Inbred BALB C ,Innate immune system ,biology ,Cell growth ,Chemistry ,CD47 ,Antigens, Differentiation ,Xenograft Model Antitumor Assays ,Immunity, Innate ,Macaca fascicularis ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Antibody - Abstract
Inhibitors of adaptive immune checkpoints have shown promise as cancer treatments. CD47 is an innate immune checkpoint receptor broadly expressed on normal tissues and overexpressed on many tumors. Binding of tumor CD47 to signal regulatory protein alpha (SIRPα) on macrophages and dendritic cells triggers a “don't eat me” signal that inhibits phagocytosis enabling escape of innate immune surveillance. Blocking CD47/SIRPα interaction promotes phagocytosis reducing tumor burden in numerous xenograft and syngeneic animal models. We have developed a next-generation humanized anti-CD47 antibody, AO-176, that not only blocks the CD47/SIRPα interaction to induce tumor cell phagocytosis, but also induces tumor cytotoxicity in hematologic and solid human tumor cell lines, but not normal noncancerous cells, by a cell autonomous mechanism (not ADCC). AO-176 also binds preferentially to tumor versus many normal cell types. In particular, AO-176 binds negligibly to RBCs in contrast to tumor cells, even at high concentrations up to 200 μg/mL and does not agglutinate RBCs up to 1 mg/mL in vitro. These properties are expected not only to decrease the antigen sink, but also to minimize on-target clinical adverse effects observed following treatment with other reported RBC-binding anti-CD47 antibodies. When tested in cynomolgus monkeys, AO-176 was well tolerated with no adverse effects. Finally, we show that AO-176 demonstrates dose-dependent antitumor activity in tumor xenograft models. Taken together, the unique properties and antitumor activity of our next-generation anti-CD47 antibody, AO-176, distinguishes it from other CD47/SIRPα axis targeting agents in clinical development.
- Published
- 2020
- Full Text
- View/download PDF
4. 260 CD47 antibody, AO-176 demonstrates potent anti-tumor activity in pre-clinical solid tumor xenografts as a single agent and in combination with multiple classes of therapeutics
- Author
-
Ronald R. Hiebsch, Robyn J Puro, Casey Wilson, Rachel Delston, Daniel S. Pereira, Arun K. Kashyap, Benjamin J. Capoccia, Michael J. Donio, and Gabriela Andrejeva
- Subjects
Pharmacology ,Cisplatin ,Cancer Research ,medicine.diagnostic_test ,Chemistry ,medicine.drug_class ,CD47 ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Monoclonal antibody ,Flow cytometry ,chemistry.chemical_compound ,Oncology ,Paclitaxel ,In vivo ,Cancer cell ,Blocking antibody ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,RC254-282 ,medicine.drug - Abstract
BackgroundCD47 is a cell surface protein expressed on tumors that binds SIRPα on macrophages and dendritic cells resulting in a ”don’t eat me” signal that allows tumors to evade phagocytosis. The highly differentiated monoclonal antibody, AO-176 directly targets CD47 and blocks this signal. AO-176 is currently being tested in phase 1 clinical trials in solid tumors and multiple myeloma. The purpose of this study was to assess in vivo efficacy of AO-176 in solid tumor models as a single agent and in combination with multiple classes of therapeutics including chemotherapeutics, monoclonal antibodies and T-cell checkpoint inhibitors.MethodsCD47 expression levels on solid tumor types were assessed by immunohistochemistry using a tumor tissue microarray. Cell-based binding was performed using flow cytometry under acidic and physiologic pH conditions to characterize the functional activity of AO-176 in the two pH environments representing tumor and normal physiologic environments. In vivo studies were performed using models of solid cancers.ResultsAll 12 solid tumor indications assessed were positive for cell membrane localized CD47 (3.3–98.6 H-scores). Cell-based binding of AO-176 to solid cancer cell lines was significantly greater (1.6–25-fold decrease in EC50, 11–39% increase in Bmax) in acidic conditions as compared to a neutral pH environment, demonstrating improved binding in the lower pH environments associated with solid tumors. AO-176 treatment in solid tumor xenograft models resulted in potent anti-tumor activity as a monotherapy (40–58% TGI) and in combination with paclitaxel in an ovarian model (99% TGI), cisplatin in an ovarian model (84% TGI), cisplatin in a gastric model (76% TGI), and an anti-VEGFR-2 in a gastric model (86% TGI). In vivo efficacy of CD47 blockade alone (~33% TGI) and in combination with anti-PD-1 (74% TGI) and anti-PD-L1 (80% TGI) T-cell checkpoint inhibitors was observed in a syngeneic model of colon cancer using a surrogate anti-CD47 blocking antibody.ConclusionsAO-176 is a differentiated anti-CD47 agent that in addition to blocking the don’t eat me signal, directly kills cancer cells, shows lower binding to normal cells such as RBCs and demonstrates increased binding activity in acidic conditions as found in the microenvironment of solid tumors. AO-176 also elicits potent anti-tumor activity in xenograft and syngeneic models as a single agent and in combination with chemotherapies, monoclonal antibodies and T-cell checkpoint inhibitors. AO-176 is currently in clinical trials as a single agent and in combination in patients with select solid cancers (NCT03834948) and in multiple myeloma (NCT04445701).
- Published
- 2021
5. 215 AO-176, a highly differentiated clinical stage anti-CD47 antibody, preferentially binds tumor versus normal cell CD47 when complexed to β1 integrin
- Author
-
John R. Richards, Daniel S. Pereira, Robyn J Puro, Arun K Kashyap, Vicki Sung, Ronald R. Hiebsch, Prabir Chakraborty, W. Casey Wilson, Michael J. Donio, Carrie Baker Brachmann, and Benjamin J. Capoccia
- Subjects
Tumor microenvironment ,Innate immune system ,biology ,medicine.diagnostic_test ,Chemistry ,Phagocytosis ,CD47 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Immune checkpoint ,Flow cytometry ,Cancer research ,biology.protein ,medicine ,Immunogenic cell death ,Antibody - Abstract
Background Overexpression of CD47 by tumor cells exploits an immune checkpoint preventing tumor recognition and destruction by innate immune cells. Binding of tumor CD47 to SIRPα on macrophages and dendritic cells triggers a ‘don’t eat me’ signal that inhibits phagocytosis and allows escape from innate immune surveillance. Blockade of the CD47/SIRPα axis, however, enables immune recognition and phagocytic clearance of tumor cells. We have developed a clinical stage CD47 targeting antibody AO-176 that is highly differentiated among agents in this class. AO-176 not only blocks the CD47/SIRPα interaction and induces phagocytosis of tumor cells, but it also has a direct killing mechanism (via PCDIII) and induction of immunogenic cell death, leveraged by preferential binding to tumor versus normal cell CD47. Methods CD47 and β1 integrin expression and localization were evaluated using a combination of flow cytometry, western blotting, confocal microscopy and immunohistochemistry. Results Previously, we described that the preferential binding of AO-176 to tumor versus normal cells was due to its interaction with CD47 molecules that were pre-complexed to β1 integrin. This finding was particularly important and suggestive of why AO-176 does not bind red blood cells since they do not express β1 integrin. We have extended these findings to show that β1 integrin as well as CD47 are also expressed at lower levels in normal versus tumor cells, and that solid and hematologic tumor cells overexpress both CD47 and β1 integrin which correlate with poor prognosis in cancer. In addition, we show that AO-176 is able to bind and occupy CD47/β1 integrin complexes to a greater extent at acidic versus physiologic pH such as would be found in tumor microenvironments, an observation that also contributes to the enhanced targeting of AO-176 to tumor cells. Taken together, these findings add further insight into the preferential binding of AO-176 to tumor versus normal cells. Conclusions The context dependent binding of AO-176 to CD47, when complexed to β1 integrin, is unique among CD47 axis targeting agents and together with its direct killing mechanism of action offers a potentially better safety profile and opportunity for a therapeutic advantage. AO-176 is currently being evaluated in Phase 1 clinical trials for the treatment of patients with select solid tumors (NCT03834948) and multiple myeloma (NCT04445701). Trial Registration NCT03834948, NCT04445701.
- Published
- 2020
6. Novel SIRPα Antibodies That Induce Single-Agent Phagocytosis of Tumor Cells while Preserving T Cells
- Author
-
Ronald R. Hiebsch, Daniel S. Pereira, Robyn J Puro, Benjamin J. Capoccia, Gabriela Andrejeva, Isra M. Darwech, and Michael J. Donio
- Subjects
THP-1 Cells ,Phagocytosis ,T cell ,T-Lymphocytes ,Immunology ,Antigen presentation ,Population ,03 medical and health sciences ,Jurkat Cells ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Neoplasms ,medicine ,Immunology and Allergy ,Macrophage ,Humans ,Receptors, Immunologic ,education ,Immunotherapy and Vaccines ,Tumor microenvironment ,education.field_of_study ,Antigen Presentation ,Innate immune system ,Chemistry ,CD47 ,Macrophages ,U937 Cells ,Antigens, Differentiation ,Cell biology ,Neoplasm Proteins ,medicine.anatomical_structure ,030215 immunology - Abstract
The signal regulatory protein α (SIRPα)/CD47 axis has emerged as an important innate immune checkpoint that enables cancer cell escape from macrophage phagocytosis. SIRPα expression is limited to macrophages, dendritic cells, and neutrophils—cells enriched in the tumor microenvironment. In this study, we present novel anti-SIRP Abs, SIRP-1 and SIRP-2, as an approach to targeting the SIRPα/CD47 axis. Both SIRP-1 and SIRP-2 bind human macrophage SIRPα variants 1 and 2, the most common variants in the human population. SIRP-1 and SIRP-2 are differentiated among reported anti-SIRP Abs in that they induce phagocytosis of solid and hematologic tumor cell lines by human monocyte-derived macrophages as single agents. We demonstrate that SIRP-1 and SIRP-2 disrupt SIRPα/CD47 interaction by two distinct mechanisms: SIRP-1 directly blocks SIRPα/CD47 and induces internalization of SIRPα/Ab complexes that reduce macrophage SIRPα surface levels and SIRP-2 acts via disruption of higher-order SIRPα structures on macrophages. Both SIRP-1 and SIRP-2 engage FcγRII, which is required for single-agent phagocytic activity. Although SIRP-1 and SIRP-2 bind SIRPγ with varying affinity, they show no adverse effects on T cell proliferation. Finally, both Abs also enhance phagocytosis when combined with tumor-opsonizing Abs, including a highly differentiated anti-CD47 Ab, AO-176, currently being evaluated in phase 1 clinical trials, NCT03834948 and NCT04445701. SIRP-1 and SIRP-2 are novel, differentiated SIRP Abs that induce in vitro single-agent and combination phagocytosis and show no adverse effects on T cell functionality. These data support their future development, both as single agents and in combination with other anticancer drugs.
- Published
- 2020
7. Abstract P1-10-15: Development of humanized anti-CD47 monoclonal antibodies with differentiated functional profiles
- Author
-
Pamela T. Manning, Robyn J Puro, S Chanda, RW Karr, Benjamin J. Capoccia, Michael J. Donio, Ronald R. Hiebsch, K. J. M. Liu, and K Crowley
- Subjects
Cancer Research ,medicine.diagnostic_test ,biology ,medicine.drug_class ,Chemistry ,CD47 ,Cell ,Monoclonal antibody ,Flow cytometry ,medicine.anatomical_structure ,Immune system ,Oncology ,Cell culture ,medicine ,biology.protein ,Cancer research ,Signal-regulatory protein alpha ,Antibody - Abstract
CD47 is a cell surface transmembrane protein that binds to signal regulatory protein alpha (SIRPα) on macrophages and results in a “don't eat me” signal that inhibits phagocytosis. Breast cancer cells, both primary and metastatic, frequently overexpress CD47 and exploit this pathway to evade macrophage-mediated destruction. Anti-CD47 monoclonal antibodies (mAbs) block the CD47/SIRPα interaction and promote tumor cell destruction via phagocytosis. Anti-CD47 mAbs also contribute to an anti-tumor T-cell response in immune-competent mice. Therefore, anti-CD47 antibodies represent a new class of immune checkpoint inhibitors that modulate both the innate and adaptive immune systems. CD47 is expressed on multiple cell types, including tumor cells and normal cells. Many anti-CD47 mAbs block the CD47/SIRPα interaction and cause phagocytosis of tumor cells, but do not directly induce the death of human tumor cells. Tioma has created new humanized anti-CD47 mAbs with novel and differentiated functional profiles to enhance functional heterogeneity. Ti-104, Ti-176 and Ti-108 block the binding of SIRPα to CD47 and increase phagocytosis of human tumor cells. They also induce cell death of human hematological and solid tumor cell lines (including breast cancer lines) in a cell autonomous manner. Cell death was determined by an increase in phosphatidylserine-positive/7AAD-positive tumor cells assessed by flow cytometry following incubation in media containing anti-CD47 mAb or a negative control immunoglobulin. In vitro, these mAbs bind to human tumor cell lines with apparent binding affinities ranging from low pM to low nM, depending on the cell line and method of analysis (solid-phase or cell-based ELISA, flow cytometry or surface plasmon resonance). In vitro, Ti-104 and Ti-108 bind to human RBCs, whereas Ti-176 has markedly reduced binding to human and cynomolgus monkey RBCs. In a four-week (once-weekly dosing) exploratory safety study in cynomolgus monkeys with Ti-176 and Ti-108, no dose-limiting toxicity or gross pathological or microscopic findings were identified after an initial dose of 5 mg/kg (Week 1) followed by doses of 50 mg/kg (Weeks 2, 3, and 4). Ti-176 treatment resulted in minimal decrease in red cell mass, hemoglobin and hematocrit, which corresponded in vitro to markedly reduced binding to cynomolgus monkey RBCs. Ti-108 caused transient reduction of RBC parameters comparable to some previously reported anti-CD47 mAbs. Ti-104, Ti-176 and Ti-108 showed potent, dose-dependent efficacy in multiple mouse tumor models, including in the MDA-MB-231 triple-negative breast cancer orthotopic model. These data provide the preclinical rationale for further evaluation of Ti-104, Ti-176 and Ti-108 as breast cancer treatments. Citation Format: Karr RW, Liu K, Hiebsch R, Capoccia B, Donio M, Crowley K, Puro R, Chanda S, Manning P. Development of humanized anti-CD47 monoclonal antibodies with differentiated functional profiles [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P1-10-15.
- Published
- 2018
- Full Text
- View/download PDF
8. AO-176, a Differentiated Clinical-Stage Anti-CD47 Antibody, Demonstrates Potent Anti-Tumor Activity across Multiple Preclinical Models of B Cell Neoplasms
- Author
-
John O. Richards, Arun K Kashyap, Mike J Donio, Daniel S. Pereira, Robyn J Puro, Gabriela Andrejeva, Ronald R. Hiebsch, Benjamin J. Capoccia, and William C. Wilson
- Subjects
Antitumor activity ,biology ,business.industry ,CD47 ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,medicine.anatomical_structure ,Cancer research ,biology.protein ,Medicine ,Stage (cooking) ,Antibody ,business ,B cell - Abstract
Upregulation of CD47, the "don't eat me" signal, on the surface of tumors to evade immune surveillance is a common escape mechanism utilized during hematological malignancy and solid tumor development, progression, and relapse. We recently reported that AO-176, a clinical stage humanized anti-CD47 IgG2 antibody, possesses differentiated characteristics such as preferential binding of tumor cells compared to normal cells, negligible binding to red blood cells, non-ADCC direct tumor killing and elicits immunogenic cell death and DAMP induction, all in addition to single-agent phagocytosis. In vivo, AO-176 has exhibited broad anti-tumor activity in preclinical xenograft models of multiple myeloma (MM), acute myeloid leukemia, T cell acute lymphoblastic leukemia, and Burkitt lymphoma. In this study, the anti-tumor activity of AO-176 in an expanded set of preclinical models of B cell neoplasms was evaluated. We assessed In vivo anti-tumor activity in a diffuse large B cell lymphoma (DLBCL) preclinical xenograft model by inoculating Toledo cells into NSG mice and treating once weekly with either 25 mg/kg AO-176 or human IgG2 isotype control. Treatment with AO-176 resulted in profound tumor shrinkage, achieved complete responses in 8/10 mice, and extended survival for all treated mice through the 46 day dosing period, compared to all isotype control treated tumors reaching endpoint by day 21. Having previously observed significant tumor shrinkage and extension of survival in subcutaneous xenograft models of MM, we sought to evaluate anti-tumor activity of AO-176 in an orthotopic model of MM. Luciferase expressing RPMI-8226 cells were inoculated via intratibial injection into NOD-SCID mice and treated with either 25 mg/kg of AO-176 or human IgG2 isotype control once weekly. AO-176-treated mice showed significant reductions of bioluminescence on study days 7, 21, 35, and 41, and serum paraprotein at study end. Evaluation of bone lesions by x-ray showed significantly reduced average bone lysis score in the AO-176 treatment group at study day 41. We then compared the anti-tumor activity of AO-176 against the second generation proteosome inhibitor carfilzomib in a myeloma xenograft model. AO-176 dosed at 25 mg/kg once weekly achieved 72% TGI, compared to 47% and 27% TGI for tumors treated with 5 mg/kg and 2.5 mg/kg carfilzomib at day 29 post treatment. To elucidate the pathways and processes that may be underpinning the anti-tumor activity of AO-176, we performed bulk RNA sequencing on AO-176 or isotype control-treated tumors harvested at multiple time points from a MM xenograft model we previously reported as exhibiting profound sensitivity to AO-176 in vivo. Murine transcripts from harvested tumor RNA were evaluated to assess differences in immune infiltrate resulting from AO-176 treatment. Days 3 and 7 post treatment with AO-176 showed the greatest number of differentially expressed genes compared to control treated tumors. The top enriched pathway on day 3 was microglia phagocytosis. Principal component analysis of gene expression indicated partitioning of day 3 post AO-176 treatment from the rest of the groups. Furthermore, deconvolution of abundances of infiltrating immune cells using CIBERSORT via TIMER analytical tool showed an enrichment of macrophages relative to other cell types on day 3 post treatment. To extend our RNA sequencing findings, we then sought to evaluate intratumoral immune cell populations after AO-176 treatment in a subcutaneous MM xenograft model. MM cells were inoculated into NOD-SCID mice, then treated with 25 mg/kg AO-176 or human IgG2 isotype control. At 48 hours post treatment, tumors were harvested, and we observed an increase of macrophages in the AO-176 treated tumors, confirming our previous results. In summary, the robust preclinical data in DLBCL and MM warrants further development of AO-176 for treatment of hematological malignancies. AO-176 is being evaluated in phase 1/2 clinical trials for the treatment of patients with solid tumors (NCT03834948) and with MM (NCT04445701). Disclosures Wilson: Arch Oncology: Current Employment. Richards: Arch Oncology: Current Employment. Andrejeva: Arch Oncology: Current Employment. Capoccia: Arch Oncology: Current Employment. Puro: Arch Oncology: Current Employment. Donio: Arch Oncology: Current Employment. Hiebsch: Arch Oncology: Current Employment. Kashyap: Arch Oncology: Current Employment. Pereira: Arch Oncology: Current Employment.
- Published
- 2021
- Full Text
- View/download PDF
9. Pre-Clinical Combination of AO-176, a Highly Differentiated Clinical Stage CD47 Antibody, with Either Azacitidine or Venetoclax Significantly Enhances DAMP Induction and Phagocytosis of Acute Myeloid Leukemia
- Author
-
Mike J Donio, Daniel S. Pereira, Robyn J Puro, Benjamin J. Capoccia, W. Casey Wilson, Isra M. Darwech, Gabriela Andrejeva, and Arun K Kashyap
- Subjects
business.industry ,Venetoclax ,T cell ,medicine.medical_treatment ,CD47 ,Immunology ,Azacitidine ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,chemistry.chemical_compound ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,chemistry ,Cancer research ,medicine ,Stem cell ,business ,medicine.drug - Abstract
While T cell checkpoint inhibitors are mainstays of cancer immunotherapy, therapies that direct innate immune responses against cancer are lacking. CD47, a "don't eat me" signal, is an innate immune cell checkpoint which binds SIRPα on macrophages and dendritic cells to limit phagocytosis and its upregulation on tumor cells leads to evasion of immune detection and clearance. Therapeutic antibodies have previously been developed to block CD47 and induce phagocytosis of tumor cells, thus validating the pathway. AO-176, a next generation humanized IgG2 anti-CD47 antibody, was developed to block the CD47/SIRPα interaction and induce tumor cell phagocytosis. Moreover, AO-176 directly kills tumor cells through a non-ADCC-dependent mechanism via induction of programmed cell death type III. In addition to these tumor eliminating properties, AO-176 has the potential for a strong safety profile as a result of its preferential binding to tumor versus normal cells, lack of RBC binding, and enhanced binding to tumor cells at acidic pH. CD47 has previously been shown to be upregulated on acute myeloid leukemia (AML) leukemic stem cells (LSC), enabling their expansion through evasion from phagocytic clearance. As a result, patients with increased CD47 on AML LSCs have worse overall survival. In this study, AO-176 efficacy was evaluated in AML cell lines as a single agent and in combination with azacitidine and venetoclax which are approved therapies for AML. Azacitidine is a cytosine analogue which acts to inhibit DNA methylation, and venetoclax is a potent Bcl-2 inhibitor. Previous studies have shown that azacitidine induces apoptosis of tumor cells and increases cell surface exposure of calreticulin, a DAMP (Damage Associate Molecular Pattern) which provides a strong pro-phagocytic signal. From these findings, it was hypothesized that azacitidine would enable increased tumor cell phagocytosis when combined with AO-176. The potential for a similar enhancement with a combination of AO-176 and venetoclax was also explored. The ability of AO-176, with or without azacitidine or venetoclax, to induce DAMPs on the surface of AML cells was assessed. Cell surface expression of DAMPs, calreticulin and PDIA3, were measured by flow cytometry. AO-176, azacitidine, and venetoclax as single agents potently increased both calreticulin and PDIA3 in a dose-dependent manner on AML cell lines such as HL60 This is the first time, to our knowledge, that venetoclax has been shown to induce DAMPs. To better understand the functional implications of these findings, in vitro phagocytosis assays were performed. Azacitidine and venetoclax significantly enhanced AO-176-mediated phagocytosis of AML cells compared to any of the agents alone. Moreover, when AO-176 was combined with azacitidine in direct tumor cell killing assays, enhanced activity was observed in a subset of AML cell lines. In conclusion, AO-176 combined with either azacitidine or venetoclax, resulted in significant enhancement of phagocytic AML cell clearance in vitro which also correlated with the ability of these agents to induce DAMPs. In vivo treatment with AO-176 in combination with these agents is in progress. AO-176 is being evaluated in phase 1 clinical trials for the treatment of patients with solid tumors (NCT03834948) and multiple myeloma (NCT04445701). Disclosures Donio: Arch Oncology: Current Employment, Current equity holder in private company. Wilson:Arch Oncology: Current Employment, Current equity holder in private company. Darwech:Arch Oncology: Current Employment, Current equity holder in private company. Andrejeva:Arch Oncology: Current Employment, Current equity holder in private company. Capoccia:Arch Oncology: Current Employment, Current equity holder in private company. Puro:Arch Oncology: Current Employment, Current equity holder in private company. Kashyap:Arch Oncology: Current Employment, Current equity holder in private company. Pereira:Arch Oncology: Current Employment, Current equity holder in private company.
- Published
- 2020
- Full Text
- View/download PDF
10. CD47 Blockade Reduces Ischemia-Reperfusion Injury and Improves Outcomes in a Rat Kidney Transplant Model
- Author
-
William A. Frazier, Joseph P. Gaut, Ronald R. Hiebsch, Chun-Cheng Chen, Benjamin J. Capoccia, Zhenyu Xiao, Pamela T. Manning, William C. Chapman, Gundumi A. Upadhya, Thalachallour Mohanakumar, Yiing Lin, and Jianluo Jia
- Subjects
Male ,medicine.medical_specialty ,Ischemia ,CD47 Antigen ,Inflammation ,Kidney ,urologic and male genital diseases ,Article ,Organ transplantation ,Nitric oxide ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,cardiovascular diseases ,Kidney transplantation ,Transplantation ,urogenital system ,business.industry ,CD47 ,Graft Survival ,fungi ,Antibodies, Monoclonal ,medicine.disease ,Kidney Transplantation ,female genital diseases and pregnancy complications ,Rats ,Blockade ,Treatment Outcome ,chemistry ,Rats, Inbred Lew ,Reperfusion Injury ,Anesthesia ,Cardiology ,medicine.symptom ,business ,Reperfusion injury - Abstract
Ischemia-reperfusion injury (IRI) significantly contributes to delayed graft function and inflammation, leading to graft loss. Ischemia-reperfusion injury is exacerbated by the thrombospondin-1-CD47 system through inhibition of nitric oxide signaling. We postulate that CD47 blockade and prevention of nitric oxide inhibition reduce IRI in organ transplantation.We used a syngeneic rat renal transplantation model of IRI with bilaterally nephrectomized recipients to evaluate the effect of a CD47 monoclonal antibody (CD47mAb) on IRI. Donor kidneys were flushed with CD47mAb OX101 or an isotype-matched control immunoglobulin and stored at 4°C in University of Wisconsin solution for 6 hr before transplantation.CD47mAb perfusion of donor kidneys resulted in marked improvement in posttransplant survival, lower levels of serum creatinine, blood urea nitrogen, phosphorus and magnesium, and less histological evidence of injury. In contrast, control groups did not survive more than 5 days, had increased biochemical indicators of renal injury, and exhibited severe pathological injury with tubular atrophy and necrosis. Recipients of CD47mAb-treated kidneys showed decreased levels of plasma biomarkers of renal injury including Cystatin C, Osteopontin, Tissue Inhibitor of Metalloproteinases-1 (TIMP1), β2-Microglobulin, Vascular Endothelial Growth Factor A (VEGF-A), and clusterin compared to the control group. Furthermore, laser Doppler assessment showed higher renal blood flow in the CD47mAb-treated kidneys.These results provide strong evidence for the use of CD47 antibody-mediated blockade to reduce IRI and improve organ preservation for renal transplantation.
- Published
- 2014
- Full Text
- View/download PDF
11. Abstract 548: Evaluation of novel SIRP antibodies as potential cancer therapeutics
- Author
-
Ronald R. Hiebsch, Myriam N. Bouchlaka, Benjamin J. Capoccia, Michael J. Donio, Prabir Chakraborty, W. Casey Wilson, Robyn J. Puro, and Daniel S. Pereira
- Subjects
Cancer Research ,Oncology - Abstract
Targeting immune checkpoints of adaptive immunity has shown great therapeutic efficacy in oncology, but in a limited fraction of patients. Innate immune cells represent the most abundant immune cell types in many solid tumors and are often linked to a poor prognosis. SIRPα is expressed by innate immune cells and its interaction with CD47, expressed by most tumor cells, is an important immune checkpoint of the innate response, involved in the regulation of phagocytosis by macrophages, dendritic cells and neutrophils. Recently, first generation agents targeting CD47 (CD47 antibodies and SIRPα-Fc fusion proteins) have shown promise in clinical trials, but they have also experienced hematological toxicities such as anemia or thrombocytopenia. Consequently, we have previously reported on the development of AO-176, a next generation anti-CD47 antibody that not only blocks the CD47/SIRPα interaction and induces phagocytosis, but also preferentially binds tumor versus normal cells (including RBCs where it binds negligibly) and directly kills tumor cells via a programmed cell death type III and an immunogenic cell death process. Here we report the discovery of novel anti-SIRP antibodies that recognize either SIRPα selectively or SIRPα/γ. These antibodies are being evaluated for their ability to induce phagocytosis of tumor cells - we have identified antibodies that induce phagocytosis of tumor cells alone and in combination with Rituxan. The ability of our anti-SIRP antibodies to induce immunomodulatory activities in a variety of ex vivo cultured immune cells expressing either SIRPα or SIRPα/γ is also under investigation and will be presented. Citation Format: Ronald R. Hiebsch, Myriam N. Bouchlaka, Benjamin J. Capoccia, Michael J. Donio, Prabir Chakraborty, W. Casey Wilson, Robyn J. Puro, Daniel S. Pereira. Evaluation of novel SIRP antibodies as potential cancer therapeutics [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 548.
- Published
- 2019
- Full Text
- View/download PDF
12. Abstract 540: AO-176, a normal cell sparing humanized anti-CD47 antibody
- Author
-
Prabir Chakraborty, Myriam N. Bouchlaka, Benjamin J. Capoccia, Ronald R. Hiebsch, Michael J. Donio, Robyn J. Puro, Vicki Sung, and Daniel S. Pereira
- Subjects
Cancer Research ,Oncology - Abstract
AO-176 is a next generation humanized anti-CD47 IgG2 that binds human and cynomolgus monkey CD47 equivalently. AO-176, like other CD47 antibodies, blocks the interaction of CD47 with SIRPα, inducing phagocytosis of tumor cells by activated macrophages. Unique to AO-176 is its ability to directly kill tumor cells via programmed cell death type III and immunogenic cell death in a cell autonomous (non-ADCC) manner. AO-176 also exhibits preferential binding to tumor vs. normal cells, a notable characteristic, as CD47 is expressed on many normal cells including red blood cells (RBC), platelets, T cells and endothelial cells. While other CD47 antibodies induce hematological toxicities such as anemia and thrombocytopenia in both primate models and in patients, AO-176 negligibly binds RBCs and platelets and even at high doses, minimally impacts hematology in cynomolgus monkey toxicology studies. AO-176 is currently being evaluated in phase 1 clinical trials for the treatment of solid tumors. In recent head to head experiments conducted with other CD47 antibodies, AO-176 bound all normal cells tested to a significantly lower degree. For example, unlike anti-CD47 antibodies such as Hu-5F9-G4, we observed negligible and minimal ex vivo binding to healthy human RBCs and platelets respectively and significantly lower binding of AO-176 to other hematologic and non-hematologic cells such as T cells and endothelial cells. Although AO-176 binds cynomolgus monkey RBCs to a slightly greater degree than human RBCs ex vivo, AO-176 versus comparative published Hu5F9-G4 findings has demonstrated dramatically reduced receptor occupancy when evaluated in monkey toxicology studies. We have also developed a clinic-ready receptor occupancy assay to measure AO-176 binding to circulating PBMCs in patients treated with AO-176 and to demonstrate minimal binding to CD47 on normal human cells. We have begun to evaluate mechanisms that may underlie the normal cell sparing effects of AO-176. Published studies have demonstrated that in addition to CD47 protein expression level, clustering and mobility of CD47 at the cell surface may impact binding to ligands, downstream signaling and subsequent cellular responses such as apoptosis. For example, avidity for SIRPα and clearance by macrophages is modulated by CD47 clustering that can be modulated by protein glycosylation, association with lipid rafts, cytoskeleton, integrins or other cis-acting factors. Here, we compare the distribution of CD47 on tumor and RBCs to evaluate receptor association with other cellular membrane components that may impact distribution on the cell surface. In summary, AO-176 is a next generation anti-CD47 antibody that, aside from induction of phagocytosis, possesses additional attributes that include direct tumor cell killing and preferential binding to tumor versus normal cells. These attributes of AO-176 differentiate it from other CD47 axis targeting agents currently in clinical evaluation. Citation Format: Prabir Chakraborty, Myriam N. Bouchlaka, Benjamin J. Capoccia, Ronald R. Hiebsch, Michael J. Donio, Robyn J. Puro, Vicki Sung, Daniel S. Pereira. AO-176, a normal cell sparing humanized anti-CD47 antibody [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 540.
- Published
- 2019
- Full Text
- View/download PDF
13. Autoimmune Gastritis Mediated by CD4+ T Cells Promotes the Development of Gastric Cancer
- Author
-
Thanh-Long M. Nguyen, Clifford J. Bellone, Russell A. Kesman, John E. Sagartz, Shradha S. Khurana, Benjamin J. Capoccia, Jason C. Mills, and Richard J. DiPaolo
- Subjects
CD4-Positive T-Lymphocytes ,Cancer Research ,Pathology ,medicine.medical_specialty ,Autoimmune Gastritis ,Blotting, Western ,Receptors, Antigen, T-Cell ,Fluorescent Antibody Technique ,Chronic gastritis ,Mice, Transgenic ,Adenocarcinoma ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,Autoimmune Diseases ,Immunoenzyme Techniques ,H(+)-K(+)-Exchanging ATPase ,Interferon-gamma ,Mice ,Stomach Neoplasms ,Metaplasia ,medicine ,Gastric mucosa ,Animals ,Humans ,RNA, Messenger ,Inflammation ,Mice, Inbred BALB C ,Reverse Transcriptase Polymerase Chain Reaction ,Stomach ,digestive, oral, and skin physiology ,Cancer ,Flow Cytometry ,medicine.disease ,digestive system diseases ,medicine.anatomical_structure ,Oncology ,Gastric Mucosa ,Gastric Polyp ,Gastritis ,Lymph Nodes ,medicine.symptom - Abstract
Chronic inflammation is a major risk factor for cancer, including gastric cancers and other gastrointestinal cancers. For example, chronic inflammation caused by autoimmune gastritis (AIG) is associated with an increased risk of gastric polyps, gastric carcinoid tumors, and possibly adenocarcinomas. In this study, we characterized the progression of gastric cancer in a novel mouse model of AIG. In this model, disease was caused by CD4+ T cells expressing a transgenic T-cell receptor specific for a peptide from the H+/K+ ATPase proton pump, a protein expressed by parietal cells in the stomach. AIG caused epithelial cell aberrations that mimicked most of those seen in progression of human gastric cancers, including chronic gastritis followed by oxyntic atrophy, mucous neck cell hyperplasia, spasmolytic polypeptide-expressing metaplasia, dysplasia, and ultimately gastric intraepithelial neoplasias. Our work provides the first direct evidence that AIG supports the development of gastric neoplasia and provides a useful model to study how inflammation drives gastric cancer. Cancer Res; 73(7); 2117–26. ©2013 AACR.
- Published
- 2013
- Full Text
- View/download PDF
14. The ubiquitin ligase Mindbomb 1 coordinates gastrointestinal secretory cell maturation
- Author
-
Richardson M Peek Jr, Matteo Fassan, Jason C. Mills, Massimo Rugge, Ramon U. Jin, Young-Yun Kong, and Benjamin J. Capoccia
- Subjects
Ubiquitin-Protein Ligases ,Cellular differentiation ,Adenocarcinoma ,Cell Enlargement ,Biology ,Cell fate determination ,Microtubules ,Mice ,Microtubule ,Intestinal Neoplasms ,Cell polarity ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Progenitor cell ,Transcription factor ,Conserved Sequence ,Mice, Knockout ,Chief Cells, Gastric ,Metaplasia ,Base Sequence ,Stomach ,Cell Polarity ,Cell Differentiation ,General Medicine ,Transport protein ,Ubiquitin ligase ,Cell biology ,Death-Associated Protein Kinases ,Protein Transport ,Tamoxifen ,Biochemistry ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Apoptosis Regulatory Proteins ,Research Article - Abstract
After cell fate specification, differentiating cells must amplify the specific subcellular features required for their specialized function. How cells regulate such subcellular scaling is a fundamental unanswered question. Here, we show that the E3 ubiquitin ligase Mindbomb 1 (MIB1) is required for the apical secretory apparatus established by gastric zymogenic cells as they differentiate from their progenitors. When Mib1 was deleted, death-associated protein kinase–1 (DAPK1) was rerouted to the cell base, microtubule-associated protein 1B (MAP1B) was dephosphorylated, and the apical vesicles that normally support mature secretory granules were dispersed. Consequently, secretory granules did not mature. The transcription factor MIST1 bound the first intron of Mib1 and regulated its expression. We further showed that loss of MIB1 and dismantling of the apical secretory apparatus was the earliest quantifiable aberration in zymogenic cells undergoing transition to a precancerous metaplastic state in mouse and human stomach. Our results reveal a mechanistic pathway by which cells can scale up a specific, specialized subcellular compartment to alter function during differentiation and scale it down during disease.
- Published
- 2013
- Full Text
- View/download PDF
15. Abstract A147: AO-176, a next-generation anti-CD47 antibody, induces immunogenic cell death
- Author
-
Daniel S. Pereira, Alun Carter, Benjamin J. Capoccia, Ronald R. Hiebsch, Michael J. Donio, Robert W. Carr, Robyn J Puro, Pamela T. Manning, and W. Casey Wilson
- Subjects
0301 basic medicine ,Cancer Research ,Innate immune system ,biology ,business.industry ,CD47 ,medicine.medical_treatment ,Immunology ,Immunotherapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Antigen ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,biology.protein ,Immunogenic cell death ,Antibody ,business - Abstract
Recent success in cancer immunotherapy has targeted immune checkpoints such as PD-1, PDL-1, and CTLA-4 to enhance the cytotoxic activity of the adaptive T-cell immune response. While the clinical response to these therapies has been dramatic for some, many others have shown partial or even no response highlighting the need for alternative or synergistic approaches that activate innate immunity. Disruption of the interaction between SIRP alpha and CD47, an innate checkpoint inhibitor, using anti-CD47 antibodies, for example, is known to enhance innate immunity by increasing the phagocytosis of tumor cells by macrophages and dendritic cells (DCs) leading to processing and presentation of tumor antigens. Recently, we described AO-176, a next generation anti-CD47 antibody that blocks the CD47/SIRP alpha interaction, induces phagocytosis and causes a direct tumor cell-autonomous death while negligibly binding RBCs. Herein, we characterize the ability of our CD47 antibodies such as AO-176 to induce immunogenic cell death (ICD) and damage-associated molecular patterns (DAMPs) in tumor cells and to potentiate chemotherapy-induced ICD/DAMPs. ICD is a process whereby an agent induces cell surface exposure and release of DAMPs from dying cells which stimulates DCs and adaptive immune responses. Tumor cells were treated in vitro with our CD47 antibodies either alone or in combination with chemotherapeutics followed by assessment of ICD/DAMPs using flow cytometry and biochemical assays. RNAseq was also performed on cells undergoing CD47 antibody mediated ICD/DAMP induction to better understand how CD47 inhibition may regulate ICD. AO-176 and other CD47 antibodies, developed by Arch Oncology, caused mitochondrial stress and loss of outer-membrane integrity, typically observed prior to cells undergoing apoptosis. In addition, CD47 antibody treatment induced a significant ER stress response at the genetic level resulting in the surface exposure of ER chaperone proteins calreticulin, Hsp90, and PDIA3. Concomitantly, our CD47 antibodies increased autophagy and JAK/STAT signaling, which resulted in both ATP and HMGB1 release, respectively. Finally, we demonstrated that in combination, our antibodies potentiated the effects of ICD/DAMP-inducing chemotherapy (e.g., doxorubicin). Here, we describe the unique ability of a specific subset of next generation CD47 antibodies, such as AO-176 to induce ICD/DAMPs. RNAseq analysis of treated cells also revealed alteration of several pathways, including those where DAMPs play a role. In summary, next-generation CD47 antibodies such as AO-176 may provide a novel approach to enhancing the current landscape of checkpoint immunotherapy by enhancing both the innate and adaptive immune responses against tumors. Citation Format: Daniel S. Pereira, Benjamin J. Capoccia, Ronald R. Hiebsch, Michael J. Donio, Alun J. Carter, Robyn J. Puro, W. Casey Wilson, Pamela T. Manning, Robert W. Carr. AO-176, a next-generation anti-CD47 antibody, induces immunogenic cell death [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A147.
- Published
- 2019
- Full Text
- View/download PDF
16. Abstract 1765: A humanized anti-CD47 monoclonal antibody that directly kills human tumor cells and has additional unique functional characteristics
- Author
-
Robyn J Puro, Myriam N. Bouchlaka, Karr Robert W, K. J. M. Liu, Benjamin J. Capoccia, Ronald R. Hiebsch, Kathleen Crowley, Pamela T. Manning, Alun Carter, and Michael J. Donio
- Subjects
0301 basic medicine ,Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,Tumor microenvironment ,biology ,Chemistry ,CD47 ,Cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Tumor progression ,Cell culture ,030220 oncology & carcinogenesis ,biology.protein ,Signal-regulatory protein alpha ,medicine ,Cancer research ,Antibody - Abstract
CD47 is a cell surface glycoprotein that interacts with signal regulatory protein alpha (SIRPα) on macrophages and dendritic cells triggering a “don't eat me” signal that inhibits phagocytosis. Many tumors evade immune surveillance by overexpressing CD47, thereby preventing their recognition by phagocytes. Blocking the interaction of SIRPα/CD47 promotes phagocytosis and tumor cell destruction leading to a reduction in tumor burden. We have developed a humanized anti-CD47 antibody, AO-176, that blocks the interaction between CD47 and SIRPα and exhibits several additional novel functional characteristics. These characteristics include the induction of cell death in multiple human tumor cell lines in a cell autonomous manner (not ADCC), assessed by an increase in phosphatidylserine/7AAD positive staining. A second novel characteristic is enhanced binding to tumor cells at acidic pH. AO-176 binds to human tumor cell lines in the high pM to low nM range at physiologic pH, however, binding is enhanced up to 20-fold at an acidic pH of 6.5. The acidic pH of the tumor microenvironment which ranges from 6.4-7.2 is characteristic of solid tumors and correlates with tumor progression and metastasis. As a result of this enhanced binding at acidic pH, AO-176 has the potential added advantage of tumor-specific targeting. A third novel characteristic exhibited by AO-176 is its selective binding to tumor cells while exhibiting reduced binding to normal cells including red blood cells (cynomolgus monkey and human), endothelial, epithelial and skeletal muscle cells. In addition to these novel characteristics, AO-176 also exhibits dose-dependent efficacy in multiple mouse tumor models. Taken together, the unique combination of functional characteristics of AO-176, including induction of cell-autonomous killing, enhanced binding to tumor cells at acidic pH, significantly reduced binding to normal cells and potent in vivo efficacy provides the preclinical rationale for further development. Citation Format: Robyn Puro, Katherine Liu, Benjamin Capoccia, Michael Donio, Ronald Hiebsch, Myriam Bouchlaka, Alun Carter, Pamela Manning, Kathleen Crowley, Robert Karr. A humanized anti-CD47 monoclonal antibody that directly kills human tumor cells and has additional unique functional characteristics [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 1765.
- Published
- 2018
- Full Text
- View/download PDF
17. Recruitment of the inflammatory subset of monocytes to sites of ischemia induces angiogenesis in a monocyte chemoattractant protein-1-dependent fashion
- Author
-
Benjamin J. Capoccia, Daniel C. Link, and Alyssa D. Gregory
- Subjects
CCR2 ,Receptors, CCR2 ,Angiogenesis ,Immunology ,Ischemia ,Fluorescent Antibody Technique ,Neovascularization, Physiologic ,Enzyme-Linked Immunosorbent Assay ,Inflammation ,Biology ,Monocytes ,Neovascularization ,Mice ,Bone Marrow ,medicine ,Animals ,Immunology and Allergy ,Therapeutic angiogenesis ,Chemokine CCL2 ,Mice, Knockout ,Monocyte ,Cell Biology ,Extracellular Mediators and Effector Molecules ,Flow Cytometry ,medicine.disease ,Adoptive Transfer ,Hindlimb ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Reperfusion ,Cancer research ,Bone marrow ,medicine.symptom - Abstract
There is accumulating evidence that delivery of bone marrow cells to sites of ischemia by direct local injection or mobilization into the blood can stimulate angiogenesis. This has stimulated tremendous interest in the translational potential of angiogenic cell population(s) in the bone marrow to mediate therapeutic angiogenesis. However, the mechanisms by which these cells stimulate angiogenesis are unclear. Herein, we show that the inflammatory subset of monocytes is selectively mobilized into blood after surgical induction of hindlimb ischemia in mice and is selectively recruited to ischemic muscle. Adoptive-transfer studies show that delivery of a small number of inflammatory monocytes early (within 48 h) of induction of ischemia results in a marked increase in the local production of MCP-1, which in turn, is associated with a secondary, more robust wave of monocyte recruitment. Studies of mice genetically deficient in MCP-1 or CCR2 indicate that although not required for the early recruitment of monocytes, the secondary wave of monocyte recruitment and subsequent stimulation of angiogenesis are dependent on CCR2 signaling. Collectively, these data suggest a novel role for MCP-1 in the inflammatory, angiogenic response to ischemia.
- Published
- 2008
- Full Text
- View/download PDF
18. Injury mediated vascular regeneration requires endothelial ER71/ETV2
- Author
-
Norifumi Urao, Masuko Ushio-Fukai, Suk Ho Bhang, Kyunghee Choi, Hong Seo Choi, Fang Liu, Tae Min Kim, Sunday S. Oladipupo, Rajendra S. Apte, David M. Ornitz, Tae-Jin Lee, Ian Pitha-Rowe, Byung-Soo Kim, Rei Nakamura, Changwon Park, Dae-Sik Lim, Benjamin J. Capoccia, Hiroyuki Miyoshi, and Dongjun Lee
- Subjects
0301 basic medicine ,Male ,Vascular Endothelial Growth Factor A ,Heterozygote ,Time Factors ,Angiogenesis ,Genetic Vectors ,Ischemia ,Neovascularization, Physiologic ,Biology ,Article ,Neovascularization ,03 medical and health sciences ,Conditional gene knockout ,medicine ,Animals ,Regeneration ,Angiogenic Proteins ,Muscle, Skeletal ,Cells, Cultured ,Skin ,Mice, Knockout ,Wound Healing ,Regeneration (biology) ,Lentivirus ,Gene Transfer Techniques ,Endothelial Cells ,Kinase insert domain receptor ,Recovery of Function ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-2 ,Choroidal Neovascularization ,Hindlimb ,Vascular endothelial growth factor B ,Mice, Inbred C57BL ,Vascular endothelial growth factor A ,Disease Models, Animal ,030104 developmental biology ,Phenotype ,Gene Expression Regulation ,Immunology ,Cancer research ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Signal Transduction ,Transcription Factors - Abstract
Objective— Comprehensive understanding of the mechanisms regulating angiogenesis might provide new strategies for angiogenic therapies for treating diverse physiological and pathological ischemic conditions. The E-twenty six (ETS) factor Ets variant 2 (ETV2; aka Ets-related protein 71) is essential for the formation of hematopoietic and vascular systems. Despite its indispensable function in vessel development, ETV2 role in adult angiogenesis has not yet been addressed. We have therefore investigated the role of ETV2 in vascular regeneration. Approach and Results— We used endothelial Etv2 conditional knockout mice and ischemic injury models to assess the role of ETV2 in vascular regeneration. Although Etv2 expression was not detectable under steady-state conditions, its expression was readily observed in endothelial cells after injury. Mice lacking endothelial Etv2 displayed impaired neovascularization in response to eye injury, wounding, or hindlimb ischemic injury. Lentiviral Etv2 expression in ischemic hindlimbs led to improved recovery of blood perfusion with enhanced vessel formation. After injury, fetal liver kinase 1 ( Flk1 ), aka VEGFR2, expression and neovascularization were significantly upregulated by Etv2 , whereas Flk1 expression and vascular endothelial growth factor response were significantly blunted in Etv2 -deficient endothelial cells. Conversely, enforced Etv2 expression enhanced vascular endothelial growth factor–mediated endothelial sprouting from embryoid bodies. Lentiviral Flk1 expression rescued angiogenesis defects in endothelial Etv2 conditional knockout mice after hindlimb ischemic injury. Furthermore, Etv2 +/− ; Flk1 +/− double heterozygous mice displayed a more severe hindlimb ischemic injury response compared with Etv2 +/ − or Flk1 +/ − heterozygous mice, revealing an epistatic interaction between ETV2 and FLK1 in vascular regeneration. Conclusions— Our study demonstrates a novel obligatory role for the ETV2 in postnatal vascular repair and regeneration.
- Published
- 2015
19. G-CSF and AMD3100 mobilize monocytes into the blood that stimulate angiogenesis in vivo through a paracrine mechanism
- Author
-
Benjamin J. Capoccia, Daniel C. Link, and Rebecca M. Shepherd
- Subjects
Benzylamines ,Receptors, CXCR4 ,Angiogenesis ,medicine.medical_treatment ,Immunology ,Ischemia ,Recombinant Granulocyte Colony-Stimulating Factor ,Cyclams ,Biochemistry ,Peripheral blood mononuclear cell ,Monocytes ,Neovascularization ,Mice ,Heterocyclic Compounds ,Granulocyte Colony-Stimulating Factor ,Paracrine Communication ,Animals ,Humans ,Medicine ,CD11b Antigen ,Neovascularization, Pathologic ,business.industry ,Monocyte ,Cell Biology ,Hematology ,medicine.disease ,Mice, Inbred C57BL ,Stem Cells in Hematology ,medicine.anatomical_structure ,Cytokine ,Cancer research ,Cytokines ,Angiogenesis Inducing Agents ,medicine.symptom ,business - Abstract
There is compelling evidence that circulating angiogenic cells exist that are able to home to sites of vascular injury and stimulate angiogenesis. However, the number of angiogenic cells in the blood is low, limiting their delivery to sites of ischemia. Treatment with certain cytokines may mobilize angiogenic cells into the blood, potentially circumventing this limitation. Herein, we show that treatment with granulocyte colony-stimulating factor (G-CSF) or AMD3100, a novel CXCR4 antagonist, significantly stimulated angiogenesis in a murine model of acute hindlimb ischemia. The kinetics of angiogenic-cell mobilization by these agents appears to be distinct, with more rapid revascularization observed in AMD3100-treated mice. Combination treatment with G-CSF and AMD3100 resulted in the earliest and most complete recovery in blood flow to the ischemic hindlimb. Adoptive transfer of mobilized blood mononuclear cells, while potently stimulating angiogenesis, did not result in the significant incorporation of donor cells into the neoendothelium. Cell-fractionation studies showed that it is the monocyte population in the blood that mediates angiogenesis in this model. Collectively, these data suggest that monocytes mobilized into the blood by G-CSF or AMD3100 stimulate angiogenesis at sites of ischemia through a paracrine mechanism.
- Published
- 2006
- Full Text
- View/download PDF
20. Antibody mediated therapy targeting CD47 inhibits tumor progression of hepatocellular carcinoma
- Author
-
Vijay Subramanian, Yiing Lin, Ronald R. Hiebsch, Zhenyu Xiao, Pamela T. Manning, Shin Lin, Babak Banan, Thalachallour Mohanakumar, William A. Frazier, Haniee Chung, Katherine C. Ott, Gundumi A. Upadhya, Benjamin J. Capoccia, and William C. Chapman
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,CD47 Antigen ,Mice, SCID ,Biology ,Article ,Metastasis ,Mice ,Phagocytosis ,Antibody Specificity ,Cell Movement ,Signal-regulatory protein alpha ,medicine ,Animals ,Humans ,neoplasms ,CD47 ,Macrophages ,Liver Neoplasms ,Antibodies, Monoclonal ,Hep G2 Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,Oncology ,Cell culture ,Tumor progression ,Hepatocellular carcinoma ,Cancer research ,biology.protein ,Antibody ,Immunostaining - Abstract
Human hepatocellular carcinoma (HCC) has a high rate of tumor recurrence and metastasis, resulting in shortened survival times. The efficacy of current systemic therapies for HCC is limited. In this study, we used xenograft tumor models to investigate the use of antibodies that block CD47 and inhibit HCC tumor growth. Immunostaining of tumor tissue and HCC cell lines demonstrated CD47 over-expression in HCC as compared to normal hepatocytes. Macrophage phagocytosis of HCC cells was increased after treatment with CD47 antibodies (CD47mAbs) that block CD47 binding to SIRPα. Further, CD47 blockade inhibited tumor growth in both heterotopic and orthotopic models of HCC, and promoted the migration of macrophages into the tumor mass. Our results demonstrate that targeting CD47 by specific antibodies has potential immunotherapeutic efficacy in human HCC.
- Published
- 2015
21. Transcription factor MIST1 in terminal differentiation of mouse and human plasma cells
- Author
-
Benjamin J. Capoccia, Jason C. Mills, Andrew J. Bredemeyer, Jeffery M. Klco, John L. Frater, and Jochen K. Lennerz
- Subjects
Lipopolysaccharides ,X-Box Binding Protein 1 ,Physiology ,Cellular differentiation ,Cell ,Plasma Cells ,Regulatory Factor X Transcription Factors ,DNA-binding protein ,Antibodies ,Leukemia, Plasma Cell ,Mice ,Genetics ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Secretion ,Promoter Regions, Genetic ,Transcription factor ,Research Articles ,Chief Cells, Gastric ,biology ,Cell Differentiation ,Intracellular Membranes ,Plasma cell neoplasm ,Flow Cytometry ,Molecular biology ,Cell biology ,Gastric chief cell ,DNA-Binding Proteins ,medicine.anatomical_structure ,Gene Expression Regulation ,biology.protein ,Syndecan-1 ,Antibody ,Transcription Factors - Abstract
Despite their divergent developmental ancestry, plasma cells and gastric zymogenic (chief) cells share a common function: high-capacity secretion of protein. Here we show that both cell lineages share increased expression of a cassette of 269 genes, most of which regulate endoplasmic reticulum (ER) and Golgi function, and they both induce expression of the transcription factors X-box binding protein 1 ( Xbp1) and Mist1 during terminal differentiation. XBP1 is known to augment plasma cell function by establishing rough ER, and MIST1 regulates secretory vesicle trafficking in zymogenic cells. We examined morphology and function of plasma cells in wild-type and Mist1−/−mice and found subtle differences in ER structure but no overall defect in plasma cell function, suggesting that Mist1 may function redundantly in plasma cells. We next reasoned that MIST1 might be useful as a novel and reliable marker of plasma cells. We found that MIST1 specifically labeled normal plasma cells in mouse and human tissues, and, moreover, its expression was also characteristic of plasma cell differentiation in a cohort of 12 human plasma cell neoplasms. Overall, our results show that MIST1 is enriched upon plasma cell differentiation as a part of a genetic program facilitating secretory cell function and also that MIST1 is a novel marker of normal and neoplastic plasma cells in mouse and human tissues.
- Published
- 2010
22. Systemic levels of G-CSF and interleukin-6 determine the angiogenic potential of bone marrow resident monocytes
- Author
-
Alyssa D. Gregory, Daniel C. Link, Benjamin J. Capoccia, and Jill R. Woloszynek
- Subjects
STAT3 Transcription Factor ,Adoptive cell transfer ,Angiogenesis ,Immunology ,Macrophage polarization ,Neovascularization, Physiologic ,Bone Marrow Cells ,Monocytes ,Neovascularization ,Mice ,Ischemia ,Granulocyte Colony-Stimulating Factor ,medicine ,Immunology and Allergy ,Animals ,Therapeutic angiogenesis ,Interleukin 6 ,biology ,Interleukin-6 ,Monocyte ,Inflammation, Extracellular Mediators, & Effector Molecules ,Cell Biology ,Adoptive Transfer ,Hindlimb ,Mice, Inbred C57BL ,medicine.anatomical_structure ,biology.protein ,Bone marrow ,medicine.symptom - Abstract
Systemic signals generated by ischemia lead to STAT3 activation and M2 polarization in bone marrow-resident monocytes. There is considerable interest in the potential of cell-based approaches to mediate therapeutic angiogenesis for acute and chronic vascular syndromes. Using a mouse model of HLI, we showed previously that adoptive transfer of a small number of donor monocytes enhanced revascularization significantly. Herein, we provide data suggesting that the BM resident monocytes sense systemic signals that influence their future functional capacity. Specifically, following induction of distant ischemia, the angiogenic capacity of BM resident monocytes is reduced markedly. We provide evidence that G-CSF and IL-6 represent such “conditioning” signals. Systemic levels of G-CSF and IL-6 are increased significantly following induction of HLI. Accordingly, BM resident monocytes from ischemic mice exhibited increased pSTAT3 and STAT3 target gene expression. Finally, G-CSFR−/− and IL-6−/− mice were resistant to the deleterious effects of ischemic conditioning on monocyte angiogenic potential. RNA expression profiling suggested that ischemia-conditioned monocytes in the BM up-regulate the well-described M2 polarization markers Chi3l4 and Lrg1. Consistent with this observation, M2-skewed monocytes from SHIP−/− mice also had impaired angiogenic capacity. Collectively, these data show that G-CSF and IL-6 provide signals that determine the angiogenic potential of BM resident monocytes.
- Published
- 2010
23. How form follows functional genomics: gene expression profiling gastric epithelial cells with a particular discourse on the parietal cell
- Author
-
Benjamin J. Capoccia, Jason C. Mills, and Won Jae Huh
- Subjects
Genetics ,Physiology ,Microarray analysis techniques ,Cellular differentiation ,Gene Expression Profiling ,Stem Cells ,Epithelial Cells ,Computational biology ,Review ,Genomics ,Biology ,Models, Biological ,Gene product ,Gene expression profiling ,Transcriptome ,medicine.anatomical_structure ,Parietal Cells, Gastric ,Gastric Mucosa ,medicine ,Epithelial Physiology ,Humans ,Cell Lineage ,Functional genomics ,Parietal cell - Abstract
The cellular composition and morphology of the stomach epithelium have been described in detail; however, the molecular mechanisms that regulate the differentiation of the various cell lineages as well as the function of mature gastric cells are far less clear. Recently, dissection of the molecular anatomy of the stomach has been boosted by the advent of functional genomics, which allows investigators to determine patterns of gene expression across virtually the entire cellular transcriptome. In this review, we discuss the impact of functional genomic studies on the understanding of gastric epithelial physiology. We show how functional genomic studies have uncovered genes that are useful as new cell lineage-specific markers of differentiation and provide new insights into cell physiology. For example, vascular endothelial growth factor B (Vegfb) has been identified as a parietal cell-specific marker that may allow parietal cells to regulate the mucosal vascular network. We also discuss how functional genomics has identified aberrantly expressed genes in disease states. Human epididymis 4 (HE4), for example, was recently identified as a metaplasia-induced gene product in mice based on microarray analysis. Finally, we will examine how analysis of higher-order patterns of gene expression can go beyond simply identifying individual genes to show how cells work as integrated systems. Specifically, we show how application of a Gene Ontology (GO) analysis of gene expression patterns from multiple tissues identifies the gastric parietal cell as an outlier, unlike other differentiated cell lineages in the stomach or elsewhere in the body.
- Published
- 2009
24. Biodegradable dendritic positron-emitting nanoprobes for the noninvasive imaging of angiogenesis
- Author
-
Steve Guillaudeu, Michael J. Welch, Ashwin Ananth, Benjamin J. Capoccia, Aviv Hagooly, Jean M. J. Fréchet, Adah Almutairi, Dana R. Abendschein, Monica Shokeen, Carolyn J. Anderson, and Raffaella Rossin
- Subjects
Male ,Dendrimers ,Angiogenesis ,Integrin ,Nanoprobe ,Neovascularization, Physiologic ,Endocytosis ,Polyethylene Glycols ,Mice ,In vivo ,Ischemia ,Animals ,Nanotechnology ,Tissue Distribution ,Beta (finance) ,Integrin alphaVbeta3 ,Multidisciplinary ,biology ,Chemistry ,Hindlimb ,Mice, Inbred C57BL ,Biochemistry ,Positron-Emission Tomography ,Physical Sciences ,biology.protein ,Biophysics ,Molecular imaging ,Oligopeptides - Abstract
A biodegradable positron-emitting dendritic nanoprobe targeted at α v β 3 integrin, a biological marker known to modulate angiogenesis, was developed for the noninvasive imaging of angiogenesis. The nanoprobe has a modular multivalent core-shell architecture consisting of a biodegradable heterobifunctional dendritic core chemoselectively functionalized with heterobifunctional polyethylene oxide (PEO) chains that form a protective shell, which imparts biological stealth and dictates the pharmacokinetics. Each of the 8 branches of the dendritic core was functionalized for labeling with radiohalogens. Placement of radioactive moieties at the core was designed to prevent in vivo dehalogenation, a potential problem for radiohalogens in imaging and therapy. Targeting peptides of cyclic arginine–glycine–aspartic acid (RGD) motifs were installed at the terminal ends of the PEO chains to enhance their accessibility to α v β 3 integrin receptors. This nanoscale design enabled a 50-fold enhancement of the binding affinity to α v β 3 integrin receptors with respect to the monovalent RGD peptide alone, from 10.40 nM to 0.18 nM IC 50 . Cell-based assays of the 125 I-labeled dendritic nanoprobes using α v β 3 -positive cells showed a 6-fold increase in α v β 3 receptor-mediated endocytosis of the targeted nanoprobe compared with the nontargeted nanoprobe, whereas α v β 3 -negative cells showed no enhancement of cell uptake over time. In vivo biodistribution studies of 76 Br-labeled dendritic nanoprobes showed excellent bioavailability for the targeted and nontargeted nanoprobes. In vivo studies in a murine hindlimb ischemia model for angiogenesis revealed high specific accumulation of 76 Br-labeled dendritic nanoprobes targeted at α v β 3 integrins in angiogenic muscles, allowing highly selective imaging of this critically important process.
- Published
- 2009
25. Abstract 2: Phosphorylation of BNIP3 is a switch between life and death in cancer cells
- Author
-
Katherine E. Liu, Julie Dimitry, Benjamin J. Capoccia, Pamela T. Manning, William A. Frazier, and Karr Robert W
- Subjects
Cancer Research ,Programmed cell death ,CD47 ,HEK 293 cells ,Cancer ,Biology ,Mitochondrion ,medicine.disease ,Jurkat cells ,Cell biology ,Oncology ,Biochemistry ,Cancer cell ,medicine ,Protein kinase A - Abstract
Antibodies vs CD47 (CD47mAbs) that block the CD47-SIRPalpha interaction promote the phagocytosis of cancer cells and have efficacy in several tumor models. A select few CD47mAbs also directly kill cancer cells by lowering cellular cAMP levels leading to mitochondrial damage and cell death. These CD47mAbs are thus referred to as “dual-function” mAbs. Activation of protein kinase A prevents CD47mAb-mediated death suggesting that phosphorylation of one or more target proteins in the cancer cell can block the death mechanism. BNIP3, a member of the BH3-only family, is induced by hypoxia and oncogenes and is necessary for induction of cell death by dual-function CD47mAbs. BNIP3 can activate autophagy, a pro-survival function, and can also induce cell death by damaging mitochondria. The role of BNIP3 in cancer is controversial and context-dependent with some cancers over-expressing BNIP3 compared to low levels of expression in normal tissue, while other cancers cannot tolerate BNIP3 expression and silence the gene, often by methylation of the BNIP3 promoter. This suggests that cancers that tolerate BNIP3 expression employ an as yet unknown mechanism to protect themselves from its toxic effects. The C-terminal transmembrane (TM) domain of BNIP3 (residues 164-184) penetrates the outer mitochondrial membrane allowing the extreme C-terminal ten residue tail of BNIP3 (RRLTTSTSTF, residues 185-194) to extend across the intermembrane space to bind OPA1 on the inner mitochondrial membrane, a key step in the death mechanism. We used a phosphosite-specific antibody to detect phosphorylation of BNIP3 immunoprecipitated from Jurkat leukemia cells at residue T188 which resides in a canonical protein kinase A site (RRLT, amino acids 185-188). Using mass spectrometry of 6His-tagged BNIP3 isolated from HEK293 cells treated with 8BrcAMP, we identified as many as 4 additional phosphorylated sites in the C-terminal tail sequence (residues 189-194, TSTSTF). We generated phosphomimetic (S/T to D) and unphosphorylated (S/T to A or N) mutations at these residues and expressed the mutant BNIP3 proteins in 293 cells. All of the BNIP3 mutants associated with mitochondria but only the phosphomimetic mutants prevented BNIP3-induced mitochondrial damage and cell death. In contrast, mutation of the phosphorylated S/T residues to unphosphorylated residues resulted in rapid and extensive cell death. Importantly, phosphomimetic C-terminal BNIP3 residues blocked cell death without preventing autophagy, providing evidence that the two roles of BNIP3 can be regulated independently. We replicated these results in one lung cancer and three breast cancer cell lines. These findings suggest that phosphorylation at the C-terminus of BNIP3 is a switch that determines the pro-survival vs pro-death effects of BNIP3. Dual-function CD47mAbs may act by dephosphorylating BNIP3 expressed in tumor cells thus unleashing its killing potential. Citation Format: William Frazier, Katherine Liu, Julie Dimitry, Benjamin Capoccia, Pamela Manning, Robert Karr. Phosphorylation of BNIP3 is a switch between life and death in cancer cells. [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 2. doi:10.1158/1538-7445.AM2015-2
- Published
- 2015
- Full Text
- View/download PDF
26. Angiogenic cells can be rapidly mobilized and efficiently harvested from the blood following treatment with AMD3100
- Author
-
Daniel C. Link, Benjamin J. Capoccia, Steven M. Devine, John F. DiPersio, Rebecca M. Shepherd, David Ingram, and Kathryn Trinkaus
- Subjects
Male ,Benzylamines ,Angiogenesis ,Anti-HIV Agents ,Clinical Trials and Observations ,Injections, Subcutaneous ,Immunology ,Population ,Neovascularization, Physiologic ,Blood Donors ,Mice, SCID ,Biology ,Cyclams ,Biochemistry ,Neovascularization ,Andrology ,Mice ,Heterocyclic Compounds ,Mice, Inbred NOD ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Humans ,Therapeutic angiogenesis ,Leukapheresis ,Progenitor cell ,education ,Cell Proliferation ,Cryopreservation ,education.field_of_study ,Stem Cells ,Endothelial Cells ,Cell Biology ,Hematology ,Hematopoietic Stem Cell Mobilization ,Endothelial stem cell ,embryonic structures ,cardiovascular system ,Female ,medicine.symptom ,Stem cell ,circulatory and respiratory physiology ,Stem Cell Transplantation - Abstract
Circulating endothelial progenitor cells (EPCs) are thought to contribute to angiogenesis following vascular injury, stimulating interest in their ability to mediate therapeutic angiogenesis. However, the number of EPCs in the blood is low, limiting endogenous repair, and a method to rapidly mobilize EPCs has not been reported. In this study, healthy donors were mobilized sequentially with the CXCR4 antagonist, AMD3100, and G-CSF. The number of EPCs and circulating angiogenic cells (CACs) in the blood and pheresis product was determined and the angiogenic capacity of each cell population assessed. Compared with baseline, treatment with AMD3100 or G-CSF increased the number of blood CACs 10.0-fold ± 4.4-fold and 8.8-fold ± 3.7-fold, respectively. The number of EPCs in the blood increased 10.2-fold ± 3.3-fold and 21.8-fold ± 5.4-fold, respectively. On a percell basis, CACs harvested from G-CSF–mobilized blood displayed increased in vivo angiogenic potential compared with AMD3100-mobilized CACs. Mobilized EPCs displayed a greater proliferative capacity than EPCs isolated from baseline blood. Both CACs and EPCs were efficiently harvested by leukapheresis. Cryopreserved CACs but not EPCs retained functional activity after thawing. These data show that AMD3100 is a potent and rapid mobilizer of angiogenic cells and demonstrate the feasibility of obtaining and storing large numbers of angiogenic cells by leukapheresis.
- Published
- 2006
27. The number of endothelial progenitor cell colonies in the blood is increased in patients with angiographically significant coronary artery disease
- Author
-
Daniel C. Link, Rebecca M. Shepherd, Richard G. Bach, Benjamin J. Capoccia, and Hasan Güven
- Subjects
medicine.medical_specialty ,Neovascularization, Physiologic ,Cell Count ,Coronary Artery Disease ,030204 cardiovascular system & hematology ,Coronary Angiography ,Endothelial progenitor cell ,Severity of Illness Index ,Blood cell ,Coronary artery disease ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Medicine ,Humans ,cardiovascular diseases ,Progenitor cell ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,business.industry ,Stem Cells ,Endothelial Cells ,medicine.disease ,humanities ,Endothelial stem cell ,medicine.anatomical_structure ,Circulatory system ,cardiovascular system ,Cardiology ,Stem cell ,Cardiology and Cardiovascular Medicine ,business ,Artery - Abstract
ObjectivesThe objective of this study was to determine whether the number of endothelial progenitor cells (EPCs) and circulating angiogenic cells (CACs) in peripheral blood was associated with the presence and severity of coronary artery disease (CAD) in patients undergoing coronary angiography.BackgroundPrevious studies have suggested an inverse relationship between levels of circulating EPCs/CACs and the presence of CAD or cardiovascular risk factors, whereas other studies have observed increased numbers of EPCs in the setting of acute ischemia. However, the criteria used to identify specific angiogenic cell subpopulations and methods of evaluating CAD varied in these studies. In the present study, we used rigorous criteria to identify EPCs and CACs in the blood of patients undergoing coronary angiography.MethodsThe number of EPCs and CACs were measured in the blood of 48 patients undergoing coronary angiography. Patients with acute coronary syndromes were excluded.ResultsCompared with patients without angiographically significant CAD, the number of EPCs was increased (1.11 ± 2.50 vs. 4.01 ± 3.70 colonies/well, p = 0.004) and the number of CACs trended higher (175 ± 137 vs. 250 ± 160 cells per mm2, p = 0.09) among patients with significant CAD. The highest levels of EPCs were isolated from patients subsequently selected for revascularization (5.03 ± 4.10 colonies/well).ConclusionsIn patients referred for coronary angiography, higher numbers of EPCs, and a trend toward higher numbers of CACs, were associated with the presence of significant CAD, and EPC number correlated with maximum angiographic stenosis severity. Endothelial progenitor cell levels were highest in patients with CAD selected for revascularization.
- Published
- 2005
28. Dual-Function Anti-CD47mAbs Induce Tumor Cell Death and Promote Phagocytosis Resulting in Enhanced in Vivo Efficacy
- Author
-
John F. DiPersio, Benjamin J. Capoccia, William A. Frazier, Karr Robert W, Ronald R. Hiebsch, Pamela T. Manning, and Michael P. Rettig
- Subjects
Innate immune system ,biology ,business.industry ,CD47 ,Immunology ,Cell Biology ,Hematology ,Acquired immune system ,Biochemistry ,Immune system ,Antigen ,Cancer cell ,biology.protein ,Cancer research ,Medicine ,Cytotoxic T cell ,Antibody ,business - Abstract
Recent success in immunomodulation of cancer has targeted immune checkpoints such as CTLA-4, PD-1 and PDL-1 to enhance adaptive immunity by stimulating production of tumor-selective, cytotoxic T cells. Anti-CD47mAbs enhance innate immunity by increasing the phagocytosis of tumor cells by macrophages leading to processing and presentation of tumor antigens to prime the adaptive T cell response. Many cancers, including hematologic cancers, up-regulate the expression of CD47 presumably to avoid immune destruction. Increased CD47 expression protects cancer cells from phagocytosis by sending a “don't eat me” signal to macrophages via SIRPalpha, an inhibitory receptor that prevents phagocytosis of CD47-bearing cells. CD47mAbs that block the CD47/SIRPalpha interaction (“blocking-only” mAbs) enhance phagocytosis of cancer cells in vitro. We have identified two CD47mAbs, Vx-1000 and Vx-1004, both of which block the CD47/SIRPalpha interaction and promote phagocytosis of tumor cells by macrophages equally well. However, Vx-1004 also has the unique property of killing cancer cells, but not normal blood cells, via a direct, cell-autonomous, cytotoxic mechanism. Therefore, Vx-1004 is a dual-function antibody. Vx-1004 selectively kills a variety of hematologic cancer cells in vitro, while Vx-1000, the blocking-only mAb, does not as assessed by annexin V staining and flow cytometry (Figure 1). In dose-response studies, cell death in leukemia cells was induced in 2 hrs by To determine if the tumor-toxic activity of Vx-1004 confers enhanced efficacy in vivo compared to Vx-1000, we compared them in two mouse hematologic cancer models: murine acute promyelocytic leukemia (APL) and B cell lymphoma (BCL). Briefly, 1x106 GFP-labeled C57BL/6 APL cells were injected IV into wild-type C57BL/6 mice that were then treated IP with 0.4 mg/kg of either Vx-1000 or Vx-1004 on the day of tumor injection and on days 3 and 6 following tumor injection, a very low dose and limited dosing regimen. On day 25, the blood of these mice was analyzed for the number of circulating APL cells. As shown in Figure 2, Vx-1000 did not significantly reduce tumor burden compared to the control group. In contrast, Vx-1004 significantly reduced tumor burden compared to controls, demonstrating greater efficacy of the dual-function CD47mAb. In addition, enhanced efficacy of Vx-1004 compared to Vx-1000 was demonstrated in BCL (Figure 3). In this model, NSG mice were injected with 1x106 murine A20 lymphoma cells subcutaneously and then treated with 0.4mg/kg/day of the CD47mAbs IP for the first five days following tumor injection. In this model, Vx-1000 also failed to inhibit tumor growth compared to controls while Vx-1004 significantly reduced tumor burden at 35 days compared to both the control and Vx-1000 groups, nearly four weeks after treatment was stopped. These data demonstrate increased anti-cancer efficacy with a dual-function CD47mAb that not only blocks the CD47/SIRPalpha interaction to increase phagocytosis of cancer cells, but also selectively kills cancer cells. These studies indicate that dual-function CD47mAbs may have better anti-tumor activity in vivo and support their use in human clinical trials. Figure 1 Figure 1. Disclosures Manning: Corvus Pharmaceutical: Employment, Equity Ownership. Capoccia:Corvus Pharmaceutical: Employment, Equity Ownership. Hiebsch:Corvus Pharmaceutical: Employment, Equity Ownership. Karr:Corvus Pharmaceutical: Employment, Equity Ownership. Frazier:Corvus Pharmaceutical: Consultancy, Equity Ownership.
- Published
- 2014
- Full Text
- View/download PDF
29. Sa1703 The E3 Ubiquitin Ligase Mindbomb 1 Regulates Secretory Vesicle Maturation in Gastric Epithelial Cell Differentiation and Metaplasia
- Author
-
Jason C. Mills and Benjamin J. Capoccia
- Subjects
Hepatology ,biology ,Chemistry ,Metaplasia ,Gastroenterology ,biology.protein ,medicine ,medicine.symptom ,Secretory Vesicle ,Ubiquitin ligase ,Cell biology ,Epithelial cell differentiation - Published
- 2013
- Full Text
- View/download PDF
30. 874 EBI3 (IL-27/IL-35) Regulates the Progression From Atrophic Gastritis to Gastric Cancer
- Author
-
John E. Sagartz, Jason C. Mills, Shradha S. Khurana, Clifford J. Bellone, Benjamin J. Capoccia, Thanh-Long M. Nguyen, Jeremy Herzog, and Richard J. DiPaolo
- Subjects
Hepatology ,business.industry ,Atrophic gastritis ,Gastroenterology ,Cancer research ,Medicine ,Cancer ,EBI3 ,business ,medicine.disease - Published
- 2013
- Full Text
- View/download PDF
31. The Role of Mindbomb 1 in Gastric Epithelial Cell Differentiation and Metaplasia
- Author
-
Benjamin J. Capoccia and Jason C. Mills
- Subjects
Hepatology ,Metaplasia ,Gastroenterology ,Cancer research ,medicine ,Biology ,medicine.symptom ,Epithelial cell differentiation - Published
- 2011
- Full Text
- View/download PDF
32. Conditioning of Monocytes in the Bone Marrow by Inflammatory Cytokines Inhibits Their Angiogenic Potential at Peripheral Sites of Ischemia
- Author
-
Alyssa D. Gregory, Daniel C. Link, and Benjamin J. Capoccia
- Subjects
Adoptive cell transfer ,biology ,business.industry ,Angiogenesis ,Immunology ,Ischemia ,Cell Biology ,Hematology ,Hindlimb ,medicine.disease ,Biochemistry ,Granulocyte colony-stimulating factor ,Proinflammatory cytokine ,medicine.anatomical_structure ,medicine ,Cancer research ,biology.protein ,Bone marrow ,Interleukin 6 ,business - Abstract
There is considerable interest in the potential of cell-based approaches to mediate therapeutic angiogenesis for a number of acute and chronic vascular syndromes. Accumulating evidence suggests that monocytes play a key role in the regulation of angiogenesis at sites of ischemia. Our laboratory has recently demonstrated that the adoptive transfer of a small number of bone marrow monocytes significantly enhanced revascularization in a murine model of acute hindlimb ischemia (Capoccia, 2008). However, the number of donor monocytes recruited to sites of ischemia was small compared to recipient (endogenous) monocytes; only 1% of the total monocytes in the ischemic muscle were of donor origin. This observation suggested that the angiogenic capacity of endogenous monocytes in the ischemic recipient was impaired. To test this hypothesis, we harvested monocytes from the bone marrow of ischemic or control mice, adoptively transferred them into recipient mice 24 hours after surgical induction of hindlimb ischemia, and measured revascularization using laser Doppler imaging. Compared with control cells, monocytes harvested from ischemic mice had markedly reduced capacity to stimulate revascularization [ratio of blood flow in ischemic versus nonischemic limb: 0.92 ± 0.04 (control) and 0.60 ± 0.10 (ischemic) on day 14; p < 0.05]. Interestingly, the ischemia-conditioned and control monocytes were recruited to the ischemic tissue equally. We next performed RNA expression profiling on monocytes harvested from the bone marrow of ischemic and control mice. The expression of a number of STAT3 target genes was induced, including SOCS3. Accordingly, using a phosphoflow assay, we observed a significant increase in STAT3 phosphorylation in ischemia-conditioned monocytes (mean fluorescent intensity: 34 ± 4) compared with control monocytes (17 ± 2; p
- Published
- 2008
- Full Text
- View/download PDF
33. Aldehyde Dehydrogenase-Activity Purifies Multiple Hemangiogenic Lineages That Accelerate Vascularization of Ischemic Tissue through Paracrine Support of Neovessel Formation
- Author
-
Sarah A. Hohm, Marian J. Neelamkavil, Benjamin J. Capoccia, Debra L. Robson, Daniel C. Link, David A. Hess, Jan A. Nolta, and Krysta Levac
- Subjects
Matrigel ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Immunology ,CD34 ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Endothelial cell differentiation ,Cell biology ,Cell therapy ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Bone marrow ,Stem cell - Abstract
In the context of regenerative therapies, the contributions of transplanted stem cells are not limited to the direct replacement of damaged cells. Easily procured, post-natal bone marrow (BM) contains several colony-forming cell (CFC) lineages that promote repair in damaged tissues through paracrine activities. However, determination of regenerative mechanisms after transplantation has proven difficult due to the involvement of multiple cell types in angiogenesis and inflammation. To enrich for human BM cells with hemangiogenic functions, we FACS purified based on low versus high aldehyde dehydrogenase (ALDH) activity, an enzyme with increased expression in hematopoietic and neural precursor cells. In contrast to ALDHlo cells which were mainly comprised of mature T- and B-cells (>90%), ALDHhi high cells were rare (8.2±1.3% vs. 0.8±0.2% of BM cells, n=8), and showed increased expression of primitive hematopoietic/endothelial (CD34, CD133, p100-fold enrichment of respective CFCs compared to unfractionated BM cells. In contrast to mature human aortic endothelial cells, secondary cultures of ECFC in matrigel matrix did not result in organized tubule formation (n=4), suggesting that ALDH-purified ECFC lacked full endothelial cell differentiation in vitro. On the contrary, ALDHhi MSCFC were highly proliferative and showed multipotent differentiation in secondary cultures for adipocyte, osteocyte, and chondrocyte formation. To assess pro-angiogenic function in vivo, we ligated the femoral artery in NOD/SCID β2M null mice, and performed i.v. transplantation without preparative irradiation within 24 hours of surgery. Compared to mice injected with unpurified BM (n=5) or ALDHlo cells (n=7), mice transplanted with ALDHhi cells (n=8) showed significantly accelerated recovery of perfusion within 7 days post-transplantation by laser Doppler perfusion imaging (p
- Published
- 2007
- Full Text
- View/download PDF
34. The Inflammatory Subset of Monocytes Stimulates Angiogenesis at Sites of Ischemia by Altering the Balance of Pro- and Anti-Inflammatory Cytokines
- Author
-
Alyssa D. Gregory, Benjamin J. Capoccia, and Daniel C. Link
- Subjects
Adoptive cell transfer ,Chemokine ,biology ,business.industry ,Angiogenesis ,medicine.medical_treatment ,Immunology ,Ischemia ,Cell Biology ,Hematology ,medicine.disease ,Revascularization ,Biochemistry ,medicine.anatomical_structure ,Cytokine ,CX3CR1 ,biology.protein ,Medicine ,Bone marrow ,business - Abstract
Our laboratory has previously shown that early delivery of monocytes to ischemic tissue by mobilization from the bone marrow to blood or by adoptive transfer enhances revascularization after acute vascular injury. There is accumulating evidence that monocytes represent a heterogeneous population, containing subsets with distinct functional properties. Herein, we show that the inflammatory subset of monocytes mediates the angiogenic response in a murine model of hindlimb ischemia induced by femoral artery excision (FAE). Inflammatory (CX3CR1loGr-1+) and resident (CX3CR1hiGr-1−) monocytes were sorted from the bone marrow of CX3CR1+/GFP mice and injected intravenously into recipient mice 24 after FAE. Revascularization was monitored by laser Doppler imaging. A marked improvement in blood flow was observed after adoptive transfer of inflammatory but not resident monocytes [ratio of blood flow in ischemic to nonischemic limb 7 days after surgery: 0.75±0.05 (inflammatory monocytes); 0.40±0.07 (resident monocytes); 0.40±0.03 (saline control); P
- Published
- 2007
- Full Text
- View/download PDF
35. The Differential Role of Stromal Derived Factor-1 (SDF-1) and Monocyte Chemoattractant Protein-1 (MCP-1) in the Recruitment of Angiogenic Cells to Ischemic Tissue
- Author
-
Benjamin J. Capoccia and Daniel C. Link
- Subjects
Adoptive cell transfer ,Chemokine ,Stromal cell ,Monocyte chemotaxis ,biology ,Angiogenesis ,business.industry ,Monocyte ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,medicine.anatomical_structure ,Cancer research ,medicine ,biology.protein ,Bone marrow ,Therapeutic angiogenesis ,business - Abstract
There is compelling evidence that circulating angiogenic cells exist in humans that are able to home to sites of vascular injury and stimulate angiogenesis. This observation has led to tremendous interest in the translational potential of these cells to mediate therapeutic angiogenesis. However, the number of angiogenic cells in the blood at baseline is low, limiting their delivery to sites of ischemia. To circumvent this limitation angiogenic cells can be mobilized into the blood. Using a murine model of acute hindlimb ischemia, we previously showed that treatment with G-CSF and/or AMD3100 potently enhanced revascularization. Adoptive transfer studies showed that mobilized monocytes mediate this response. Finally, the monocytes, once recruited to the ischemic tissue, stimulate angiogenesis in a paracrine fashion. The signals that recruit monocytes to sites of ischemia remain poorly defined. A better understanding of these signals may provide important insights into strategies to enhance or inhibit angiogenic cell recruitment. Herein, we summarize our recent findings suggesting that MCP-1 and SDF-1 play key but distinct roles in this process. SDF-1, through its major receptor CXCR4, is thought to play an important role in angiogenic cell recruitment and/or retention at sites of ischemia. To test this hypothesis, we studied the angiogenic capacity of CXCR4−/− cells. Since CXCR4 deficiency is embryonic lethal, we first generated bone marrow chimeras by transplanting CXCR4−/− fetal liver cells into irradiated syngeneic mice. Hematopoietic reconstitution with CXCR4−/− cells was confirmed by flow cytometry. Endogenous revascularization following the induction of hindlimb ischemia in these CXCR4−/− chimeric mice was comparable to wild type chimeric mice. To further assess the angiogenic capacity of CXCR4−/− cells, adoptive transfer experiments into wild type recipient mice were performed. As reported previously, adoptive transfer of wild type bone marrow cells markedly improved blood flow following induction of hindlimb ischemia [ratio of ischemic to nonischemic limb perfusion on day 14 after surgery: 0.54 ± 0.04 (saline control) vs. 0.81 ± 0.06 (WT bone marrow); p MCP-1 is a major chemokine regulating monocyte chemotaxis and activation. Thus, we repeated the adoptive transfer experiments using MCP-1 deficient recipient mice. Adoptive transfer of wild type bone marrow cells into these mice had no effect on revascularization. Moreover, treatment of MCP-1−/− mice with G-CSF and AMD3100, though inducing the expected level of monocyte mobilization, failed to stimulate angiogenesis. These data highlight the importance of MCP-1 to the recruitment, retention, and/or activation of angiogenic cells at sites of ischemia.
- Published
- 2006
- Full Text
- View/download PDF
36. AMD3100 and G-CSF Mobilize Angiogenic Cells into the Blood of Healthy Donors
- Author
-
Rebecca M. Shepherd, David A. Ingram, Steven M. Devine, Daniel C. Link, and Benjamin J. Capoccia
- Subjects
CD31 ,Angiogenesis ,business.industry ,Plerixafor ,Immunology ,Cell Biology ,Hematology ,Leukapheresis ,Pharmacology ,Colony-stimulating factor ,Biochemistry ,Peripheral blood mononuclear cell ,Transplantation ,Medicine ,Progenitor cell ,business ,medicine.drug - Abstract
There is evidence that angiogenic cells exist in the blood that are able to home to sites of ischemia and induce angiogenesis. Angiogenic cell populations identified in human blood include endothelial progenitor cells (EPC) and circulating angiogenic monocytes (CAM). Under basal conditions, however, the number of angiogenic cells in the circulation is small, potentially limiting their delivery to sites of ischemia and subsequent stimulation of angiogenesis. To circumvent this limitation, animal studies have shown that angiogenic cells can be mobilized into blood by certain cytokines; whether a similar mobilization of angiogenic cells can be achieved in humans is not clear. Herein, we report the results of a clinical trial that examined the ability of G-CSF and AMD3100 to mobilize EPC and CAM into the blood. Donors were treated with a single injection of AMD3100 (5mg/kg) after which pheresis was performed. One week later, donors were treated with G-CSF (250microg/kg/d x 5 days) and pheresed. Blood was collected at baseline (prior to the initiation of the mobilizing regimen) and after treatment with AMD3100 or G-CSF (at the time of peak HSC mobilization). In addition, pheresis product was collected after mobilization by AMD3100 or G-CSF. Mononuclear cells (MNC) were isolated from the blood products and cultured under angiogenic conditions. EPC were identified by the formation of discrete colonies of endothelial cells on days 14-28. A novel method to quantify CAM was developed to avoid the pitfalls of counting adherent cells in culture. Briefly, adherent cells were recovered from the culture, counted, and analyzed by flow cytometry. CAM were identified as CD45+ CD14+ CD31+ cells. The angiogenic potential of the CAM was assessed by their transplantation into NOD/SCID/b2M−/− mice following surgical induction of hindlimb ischemia. Indeed, reperfusion was significantly enhanced in mice that received CAM compared to control mice [ratio±SEM) of ischemic to non-ischemic limb perfusion: 0.47±0.05 (saline treated); 0.78±0.09 (CAM transplant); p
- Published
- 2005
- Full Text
- View/download PDF
37. Bone Marrow-Derived Aldehyde Dehdrogenase Expressing Cells Possess Endothelial Progenitor Function in Addition to Hematopoietic Repopulating Ability and Aid in Blood Flow Recovery after Acute Ischemic Injury
- Author
-
Rebecca M. Shepherd, Jan A. Nolta, Daniel C. Link, David A. Hess, Kevin O’Dell, Louisa Wirthlin, and Benjamin J. Capoccia
- Subjects
Endothelium ,Chemistry ,Monocyte ,Immunology ,CD34 ,Cell Biology ,Hematology ,Biochemistry ,Molecular biology ,Endothelial stem cell ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Bone marrow ,Progenitor cell ,Stem cell - Abstract
Human bone marrow (BM) is a heterogeneous compartment of stem and progenitor cells from hematopoietic, endothelial, and mesenchymal lineages. These cell types may be recruited to sites of new blood vessel formation including ischemic tissues and tumor microenvironments. Prospective isolation of cells with neoangiogenic function has proven difficult due to the lack of surface markers specific to endothelial precursors. We have previously characterized a novel population of reconstituting hematopoietic stem and progenitor cells from human umbilical cord blood by selection of cells with high aldehyde dehydrogenase (ALDH) enzyme activity (Hess, Blood 2004). In the current study, we isolated ALDHhi and ALDHlo cells with low side scatter properties from human BM, and assayed for hematopoietic and angiogenic function. The ALDHhi subset represented 0.8±0.2% of total nucleated BM cells whereas ALDHlo cells were 10-fold more abundant (8.2±1.3%, n=5). ALDHhi cells highly expressed cell surface molecules associated with hematopoietic (89.1±1.3% CD45+, 70.5±7.5% CD117+) and endothelial progenitor function (76.5±1.8 CD34+, 72.0±7.6% CD133+). ALDHhi cells also expressed mature macrophage/monocyte markers (24.2±3.9% CD11b+), whereas mature lymphocyte markers (3.5±0.5% CD3+, 5.1±0.3% CD19+), and mature endothelial cell markers (2.6±0.8% VEGFR2+) were rare. The corresponding ALDHlo population demonstrated a significantly reduced frequency of cells expressing a primitive phenotype (8.6±1.5% CD34+, 1.4±0.4% CD117+, 2.2±0.6% CD133+, p
- Published
- 2005
- Full Text
- View/download PDF
38. A Comparison of the Ability of AMD3100 Versus G-CSF to Induce Angiogenesis and Mobilize Endothelial Progenitor Cells (EPCs)
- Author
-
Benjamin J. Capoccia, Daniel C. Link, Steven M. Devine, Rebecca M. Shepherd, and Hasan Güven
- Subjects
business.industry ,Angiogenesis ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Revascularization ,Biochemistry ,Transplantation ,Andrology ,Haematopoiesis ,Limb perfusion ,Medicine ,Progenitor cell ,Stem cell ,business ,Perfusion - Abstract
There is emerging evidence that circulating EPCs are able to home to sites of vascular injury and facilitate revascularization. However, under normal conditions, the number of EPCs in the blood is small, potentially limiting revascularization following acute injury. Previous reports suggest that certain cytokines including G-CSF can mobilize EPCs into the blood, potentially circumventing this limitation. However, the kinetics and magnitude of EPC mobilization by G-CSF have not been carefully defined and the angiogenic capacity of mobilized EPCs unclear. Herein, we characterize the ability of G-CSF treatment to improve revascularization in a mouse model of acute hindlimb ischemia. Since it is possible that EPC mobilization by G-CSF is delayed (maximal mobilization of hematopoietic stem cells (HSC) occurs after 4-5 days), we also studied AMD3100, a CXCR4 antagonist, that has been shown to lead to rapid and robust mobilization of HSC. Four groups of mice (n=7–10, each) were treated with saline alone, G-CSF (250μg/kg per day for 7 days by miniosmotic pump), AMD3100 (5mg/kg SQ on days 1–3), or a combination of both G-CSF and AMD3100; treatment was initiated immediately after induction of unilateral hindlimb ischemia. Revascularization was monitored by laser Doppler imaging and the ratio of perfusion between ischemic and non-ischemic calculated. At 2 weeks post surgery, a significant improvement in ischemic limb perfusion was observed in G-CSF and G-CSF+AMD3100 treated mice compared with saline treated mice with a trend towards improved perfusion noted in the AMD3100 treated mice [ratio of ischemic vs. non-ischemic limb perfusion + SEM: 71.9 ± 5.2% (saline treated); 89.2 ± 4.7%, p As described more completely in a separate abstract, we have initiated a phase II study to compare the safety and efficacy of AMD3100 versus G-CSF for mobilization and transplantation of HSC for HLA-matched sibling allografting. Donors are sequentially mobilized with AMD3100 and G-CSF. To compare the ability of AMD3100 and G-CSF to mobilize EPCs, we have begun to quantify the number of EPCs in the blood at baseline and after treatment with AMD3100 or G-CSF. A culture-based method was used to identify both early outgrowth EPCs (scored on day 7; predominantly CD14+, CD31+, monocytic cells) and late outgrowth EPCs (scored on day 14–21; predominantly CD45−, CD14−, CD31+ endothelial cells). To date only a single patient has been analyzed completely. Interestingly, treatment with G-CSF leads to a more robust mobilization of early EPCs (fold increase compared with baseline: 4.7-fold and 25-fold for AMD3100 and G-CSF, respectively). In contrast, treatment with AMD3100 leads to a more robust mobilization of late EPC. Surprisingly, no late outgrowth EPC were detected in 40 ml of baseline or G-CSF treated blood, whereas 26 were detected in 40 ml of AMD3100 treated blood. Studies are underway to confirm these findings with additional donors. Collectively, these data show that treatment with AMD3100 alone or in combination with G-CSF leads to enhanced revascularization following acute vascular injury and the rapid mobilization of EPCs.
- Published
- 2004
- Full Text
- View/download PDF
39. Transcription factor MIST1 in terminal differentiation of mouse and human plasma cells.
- Author
-
Benjamin J. Capoccia
- Subjects
- *
TRANSCRIPTION factors , *LABORATORY mice , *CELL differentiation , *PLASMA cells , *GENE expression , *ENDOPLASMIC reticulum , *CELL physiology , *CELL morphology - Abstract
Despite their divergent developmental ancestry, plasma cells and gastric zymogenic (chief) cells share a common function: high-capacity secretion of protein. Here we show that both cell lineages share increased expression of a cassette of 269 genes, most of which regulate endoplasmic reticulum (ER) and Golgi function, and they both induce expression of the transcription factors X-box binding protein 1 (Xbp1) and Mist1 during terminal differentiation. XBP1 is known to augment plasma cell function by establishing rough ER, and MIST1 regulates secretory vesicle trafficking in zymogenic cells. We examined morphology and function of plasma cells in wild-type and Mist1–/–mice and found subtle differences in ER structure but no overall defect in plasma cell function, suggesting that Mist1 may function redundantly in plasma cells. We next reasoned that MIST1 might be useful as a novel and reliable marker of plasma cells. We found that MIST1 specifically labeled normal plasma cells in mouse and human tissues, and, moreover, its expression was also characteristic of plasma cell differentiation in a cohort of 12 human plasma cell neoplasms. Overall, our results show that MIST1 is enriched upon plasma cell differentiation as a part of a genetic program facilitating secretory cell function and also that MIST1 is a novel marker of normal and neoplastic plasma cells in mouse and human tissues. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
40. How form follows functional genomics: gene expression profiling gastric epithelial cells with a particular discourse on the parietal cell.
- Author
-
Benjamin J. Capoccia
- Subjects
GENE expression ,CYTOGENETICS ,EPITHELIAL cells ,STOMACH ,ANATOMY ,CELL physiology ,VASCULAR endothelial growth factors ,MOLECULAR genetics - Abstract
The cellular composition and morphology of the stomach epithelium have been described in detail; however, the molecular mechanisms that regulate the differentiation of the various cell lineages as well as the function of mature gastric cells are far less clear. Recently, dissection of the molecular anatomy of the stomach has been boosted by the advent of functional genomics, which allows investigators to determine patterns of gene expression across virtually the entire cellular transcriptome. In this review, we discuss the impact of functional genomic studies on the understanding of gastric epithelial physiology. We show how functional genomic studies have uncovered genes that are useful as new cell lineage-specific markers of differentiation and provide new insights into cell physiology. For example, vascular endothelial growth factor B (Vegfb) has been identified as a parietal cell-specific marker that may allow parietal cells to regulate the mucosal vascular network. We also discuss how functional genomics has identified aberrantly expressed genes in disease states. Human epididymis 4 (HE4), for example, was recently identified as a metaplasia-induced gene product in mice based on microarray analysis. Finally, we will examine how analysis of higher-order patterns of gene expression can go beyond simply identifying individual genes to show how cells work as integrated systems. Specifically, we show how application of a Gene Ontology (GO) analysis of gene expression patterns from multiple tissues identifies the gastric parietal cell as an outlier, unlike other differentiated cell lineages in the stomach or elsewhere in the body. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.