Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Immunother Cancer ; 12(7)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39038918

ABSTRACT

BACKGROUND: Toll-like receptor 9 (TLR9) agonists induce inflammatory responses that promote the killing of infectious micro-organisms, cancer cells and develop adaptive immune responses. Their ability as immunomodulators to enhance the activity of checkpoint inhibitors (CPI) in treating liver tumors is limited in part by the distinctive biology of intrahepatic myeloid-derived suppressor cells (MDSC) and challenges with tumor-specific therapeutic delivery. We have shown that the regional delivery of type C TLR9 agonist via pressure-enabled drug delivery (PEDD) system improves delivery to the tumor, enhances depletion of MDSCs and overall, stimulates the immune system in combination with or without CPI. Currently, CPIs are delivered intravenously, although there is a growing interest in its subcutaneous (SQ) administration. We compared nelitolimod formerly known as SD-101 administered using PEDD in combination with systemic (Sys) or SQ CPI in murine liver metastases (LM). METHODS: The LM model was developed by injecting MC38-Luc cells via the spleen of 8-12 week old male C57/BL6 mice followed by splenectomy. After a week, fluorescently labeled nelitolimod (10 µg/mouse) was delivered via PEDD and co-administered anti-programmed cell death-1 (α-PD-1) either via Sys or SQ. Tumor burden was monitored by in vivo imaging system. Serum cytokine levels were analyzed by Luminex. Tissues were harvested on Day 3 (D3) or Day 10 (D10) post-PEDD to enrich CD45+ cells and were analyzed via NanoString targeted transcriptomics (D3) or flow cytometry (FC, D10) to interrogate immune cell populations (D10). For NanoString analysis, the innate immune panels were selected, and for FC, MDSCs (CD11b+Gr1+), B cells (B220+), dendritic cells (DC, CD11c+), T (CD3+) cells, and M1-like macrophages (F4/80+CD38+Egr2-) were quantified. RESULTS: Nelitolimod delivered via PEDD resulted in changes in innate and adaptive immune cells within LM, including depletion of liver MDSC and increased M1-like macrophages in the liver, which are supportive of antitumor immunity. While CPI monotherapy failed to control tumor progression, nelitolimod and CPI combination improved LM control, survival and antitumor immunity beyond the nelitolimod monotherapy effect, irrespective of CPI delivery route. CONCLUSION: The SQ route of CPI delivery was equivalent to Sys in combination with nelitolimod, suggesting SQ-CPI may be a rational choice in combination with PEDD of nelitolimod for liver tumor treatment.


Subject(s)
Immune Checkpoint Inhibitors , Liver Neoplasms , Myeloid-Derived Suppressor Cells , Animals , Mice , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/therapeutic use , Humans , Drug Delivery Systems , Mice, Inbred C57BL , Cell Line, Tumor
2.
Gut ; 72(2): 325-337, 2023 02.
Article in English | MEDLINE | ID: mdl-35705369

ABSTRACT

OBJECTIVE: Programmed cell death protein 1 (PD-1) checkpoint inhibition and adoptive cellular therapy have had limited success in patients with microsatellite stable colorectal cancer liver metastases (CRLM). We sought to evaluate the effect of interleukin 10 (IL-10) blockade on endogenous T cell and chimeric antigen receptor T (CAR-T) cell antitumour function in CRLM slice cultures. DESIGN: We created organotypic slice cultures from human CRLM (n=38 patients' tumours) and tested the antitumour effects of a neutralising antibody against IL-10 (αIL-10) both alone as treatment and in combination with exogenously administered carcinoembryonic antigen (CEA)-specific CAR-T cells. We evaluated slice cultures with single and multiplex immunohistochemistry, in situ hybridisation, single-cell RNA sequencing, reverse-phase protein arrays and time-lapse fluorescent microscopy. RESULTS: αIL-10 generated a 1.8-fold increase in T cell-mediated carcinoma cell death in human CRLM slice cultures. αIL-10 significantly increased proportions of CD8+ T cells without exhaustion transcription changes, and increased human leukocyte antigen - DR isotype (HLA-DR) expression of macrophages. The antitumour effects of αIL-10 were reversed by major histocompatibility complex class I or II (MHC-I or MHC-II) blockade, confirming the essential role of antigen presenting cells. Interrupting IL-10 signalling also rescued murine CAR-T cell proliferation and cytotoxicity from myeloid cell-mediated immunosuppression. In human CRLM slices, αIL-10 increased CEA-specific CAR-T cell activation and CAR-T cell-mediated cytotoxicity, with nearly 70% carcinoma cell apoptosis across multiple human tumours. Pretreatment with an IL-10 receptor blocking antibody also potentiated CAR-T function. CONCLUSION: Neutralising the effects of IL-10 in human CRLM has therapeutic potential as a stand-alone treatment and to augment the function of adoptively transferred CAR-T cells.


Subject(s)
Carcinoma , Colorectal Neoplasms , Interleukin-10 , Liver Neoplasms , Receptors, Chimeric Antigen , Receptors, Interleukin-10 , Animals , Humans , Mice , Carcinoembryonic Antigen/immunology , Carcinoma/immunology , Carcinoma/secondary , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/pathology , Immunotherapy, Adoptive , Interleukin-10/antagonists & inhibitors , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lymphocyte Activation , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Receptors, Interleukin-10/antagonists & inhibitors , Antibodies, Blocking/immunology
3.
Cancer Gene Ther ; 29(12): 1854-1865, 2022 12.
Article in English | MEDLINE | ID: mdl-35697801

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) expand in response to malignancy and suppress responsiveness to immunotherapy, including checkpoint inhibitors (CPIs). Within the liver, MDSCs have unique immunosuppressive features. While TLR9 agonists have shown promising activities in enhancing CPI responsiveness in superficial tumors amenable to direct needle injection, clinical success for liver tumors with TLR9 agonists has been limited by delivery challenges. Here, we report that regional intravascular infusion of ODN2395 into mice with liver metastasis (LM) partially eliminated liver MDSCs and reprogrammed residual MDSC. TLR9 agonist regional infusion also induced an increase in the M1/M2 macrophage ratio. Enhanced TLR9 signaling was demonstrated by an increased activation of in NFκB (pP65) and production of IL6 compared with systemic infusion. Further, PBMC-derived human MDSCs express TLR9, and treatment with class C TLR9 agonists (ODN2395 and SD101) reduced the expansion of MDSC population. TLR9 stimulation induced MDSC apoptosis and increased the M1/M2 macrophage ratio. Regional TLR9 agonist infusion along with systemic anti-PD-1 therapy improved control of LM. With effective delivery, TLR9 agonists have the potential to favorably reprogram the liver TME through reduction of MDSCs and favorable macrophage polarization, which may improve responsiveness to systemic CPI therapy.


Subject(s)
Liver Neoplasms , Myeloid-Derived Suppressor Cells , Toll-Like Receptor 9 , Animals , Humans , Mice , Cell Line, Tumor , Leukocytes, Mononuclear , Liver Neoplasms/drug therapy , Toll-Like Receptor 9/agonists , Tumor Microenvironment
4.
Biomedicines ; 10(3)2022 Mar 11.
Article in English | MEDLINE | ID: mdl-35327456

ABSTRACT

With the advent of cancer immunotherapy, there has been a major improvement in patient's quality of life and survival. The growth of cancer immunotherapy has dramatically changed our understanding of the basics of cancer biology and has altered the standards of care (surgery, radiotherapy, and chemotherapy) for patients. Cancer immunotherapy has generated significant excitement with the success of chimeric antigen receptor (CAR) T cell therapy in particular. Clinical results using CAR-T for hematological malignancies have led to the approval of four CD19-targeted and one B-cell maturation antigen (BCMA)-targeted cell therapy products by the US Food and Drug Administration (FDA). Also, immune checkpoint inhibitors such as antibodies against Programmed Cell Death-1 (PD-1), Programmed Cell Death Ligand-1 (PD-L1), and Cytotoxic T-Lymphocyte-Associated Antigen 4 (CTLA-4) have shown promising therapeutic outcomes and long-lasting clinical effect in several tumor types and patients who are refractory to other treatments. Despite these promising results, the success of cancer immunotherapy in solid tumors has been limited due to several barriers, which include immunosuppressive tumor microenvironment (TME), inefficient trafficking, and heterogeneity of tumor antigens. This is further compounded by the high intra-tumoral pressure of solid tumors, which presents an additional challenge to successfully delivering treatments to solid tumors. In this review, we will outline and propose specific approaches that may overcome these immunological and physical barriers to improve the outcomes in solid tumor patients receiving immunotherapies.

5.
Methods Cell Biol ; 167: 203-226, 2022.
Article in English | MEDLINE | ID: mdl-35152997

ABSTRACT

Chimeric Antigen Receptor T cell (CAR-T) therapies have demonstrated promising clinical response rates for patients with hematological malignancies. CAR-T cells are generated by bioengineering patient T cells to express CAR proteins that can specifically target tumor antigens. For CAR-T therapy to be effective in solid tumors, several factors including trafficking, activation, expansion, and persistence are important considerations in hostile tumor microenvironment (TME). Despite significant advances in CAR-T therapies, clinical success in solid tumor indications has been limited. The complex processes involved in CAR-T manufacturing and quality control (QC) contribute to high cost of these therapies, creating barriers to widespread adoption. The processes involved are not uniformly standardized and may require numerous manual handling steps. This chapter details strategies to overcome barriers to broad CAR-T application and reviews current production processes used for CAR-T manufacturing. Emphasis is placed on reproducibility, manufacturing costs, and product release testing. Implementing new processing alternatives and innovative CAR-T designs, along with further innovation and efficiencies will be key to more consistent CAR-T cell therapy success.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Reproducibility of Results , T-Lymphocytes/metabolism , Tumor Microenvironment
6.
J Surg Res ; 272: 37-50, 2022 04.
Article in English | MEDLINE | ID: mdl-34929499

ABSTRACT

BACKGROUND: Effective treatment of solid tumors requires multi-modality approaches. In many patients with stage IV liver disease, current treatments are not curative. Chimeric antigen receptor T cells (CAR-T) are an intriguing option following success in hematological malignancies, but this has not been translated to solid tumors. Limitations include sub-optimal delivery and elevated interstitial fluid pressures. We developed a murine model to test the impact of high-pressure regional delivery (HPRD) on trafficking to liver metastases (LM) and tumor response. MATERIALS AND METHODS: CAR-T were generated from CD45.1 mice and adoptively transferred into LM-bearing CD45.2 mice via regional or systemic delivery (RD, SD). Trafficking, tumor growth, and toxicity were evaluated with flow cytometry, tumor bioluminescence (TB, photons/sec log2-foldover baseline), and liver function tests (LFTs). RESULTS: RD of CAR-T was more effective at controlling tumor growth versus SD from post-treatment days (PTD) 2-7 (P = 0.002). HPRD resulted in increased CAR-T penetration versus low-pressure RD (LPRD, P = 0.004), suppression of tumor proliferation (P = 0.03), and trended toward improved long-term control at PTD17 (TB=3.7 versus 6.1, P = 0.47). No LFT increase was noted utilizing HPRD versus LPRD (AST/ALT P = 0.65/0.84) while improved LFTs in RD versus SD groups suggested better tumor control (HPRD AST/ALT P = 0.04/0.04, LPRD AST/ALT P = 0.02/0.02). CONCLUSIONS: Cellular immunotherapy is an emerging option for solid tumors. Our model suggests RD and HPRD improved CAR-T penetration into solid tumors with improved short-term tumor control. Barriers associated with SD can be overcome using RD techniques to maximize therapeutic delivery and HPRD may further augment efficacy without increased toxicity.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Neoplasms , Receptors, Chimeric Antigen , Animals , Colorectal Neoplasms/therapy , Humans , Immunotherapy, Adoptive/methods , Liver Neoplasms/pathology , Mice , Neoplasms/therapy , T-Lymphocytes
7.
Cell Death Discov ; 7(1): 232, 2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34482371

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) promote immunosuppressive activities in the tumor microenvironment (TME), resulting in increased tumor burden and diminishing the anti-tumor response of immunotherapies. While primary and metastatic tumors are typically the focal points of therapeutic development, the immune cells of the TME are differentially programmed by the tissue of the metastatic site. In particular, MDSCs are programmed uniquely within different organs in the context of tumor progression. Given that MDSC plasticity is shaped by the surrounding environment, the proteomes of MDSCs from different metastatic sites are hypothesized to be unique. A bottom-up proteomics approach using sequential window acquisition of all theoretical mass spectra (SWATH-MS) was used to quantify the proteome of CD11b+ cells derived from murine liver metastases (LM) and lung metastases (LuM). A comparative proteomics workflow was employed to compare MDSC proteins from LuM (LuM-MDSC) and LM (LM-MDSC) while also elucidating common signaling pathways, protein function, and possible drug-protein interactions. SWATH-MS identified 2516 proteins from 200 µg of sample. Of the 2516 proteins, 2367 have matching transcriptomic data. Upregulated proteins from lung and liver-derived murine CD11b+ cells with matching mRNA transcriptomic data were categorized based on target knowledge and level of drug development. Comparative proteomic analysis demonstrates that liver and lung tumor-derived MDSCs have distinct proteomes that may be subject to pharmacologic manipulation.

8.
Vaccines (Basel) ; 9(8)2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34451932

ABSTRACT

Metastatic liver tumors have presented challenges with the use of checkpoint inhibitors (CPIs), with only limited success. We hypothesize that regional delivery (RD) of CPIs can improve activity in the liver and minimize systemic exposure, thereby reducing immune-related adverse events (irAE). Using a murine model of colorectal cancer liver metastases (LM), we confirmed high levels of PD-L1 expression on the tumor cells and liver myeloid-derived suppressor cells (L-MDSC). In vivo, we detected improved LM response at 3 mg/kg on PTD7 via portal vein (PV) regional delivery as compared to 3 mg/kg via tail vein (TV) systemic delivery (p = 0.04). The minimal effective dose at PTD7 was 5 mg/kg (p = 0.01) via TV and 0.3 mg/kg (p = 0.02) via PV. We detected 6.7-fold lower circulating CPI antibody levels in the serum using the 0.3 mg/kg PV treatment compared to the 5 mg/kg TV cohort (p < 0.001) without increased liver toxicity. Additionally, 3 mg/kg PV treatment resulted in increased tumor cell apoptotic signaling compared to 5 mg/kg TV (p < 0.05). Therefore, RD of an anti-PD-1 CPI therapy for CRCLM may improve the therapeutic index by reducing the total dose required and limiting the systemic exposure. These advantages could expand CPI indications for liver tumors.

9.
Oncogene ; 40(3): 693-704, 2021 01.
Article in English | MEDLINE | ID: mdl-33230244

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that proliferate in the setting of cancer and have potent immunosuppressive functions hindering anti-tumor immunity. Here we establish that the immunologic landscape and tumor microenvironments (TME) vary between different organs which discretely shape MDSC repertoires. We found that pSTAT3 signaling exerts a dominant effect on MDSC programming in liver metastasis (LM). In contrast, in lung metastasis (LuM), MDSC programming is driven mainly by pSTAT5. Adoptive transfer of LM-MDSC into LuM resulted in a shift from pSTAT3 signaling to pSTAT5, in association with an overall shift toward lung MDSC programming. A shift from more immunosuppressive M-MDSC to G-MDSC, along with enhanced differentiation of MDSCs into pro-inflammatory M1 macrophages in LuM, indicated that MDSC plasticity and differentiation patterns are environmentally dependent. Using mass spectroscopy, we confirmed that LM-MDSCs showed enhanced expression of key proliferation pathway markers. This confirmed that liver-specific MDSC programing was comprehensive but reversible, implying that therapeutic targeting of LM-MDSC could prime the TME in a favorable manner. Our data suggest that MDSC programming in response to malignancy is highly dependent on organ-specific conditions and is modifiable.


Subject(s)
Granulocytes/metabolism , Lung Neoplasms/metabolism , Monocytes/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Tumor Microenvironment , Animals , Cell Line, Tumor , Granulocytes/pathology , Lung Neoplasms/pathology , Male , Mice , Monocytes/pathology , Myeloid-Derived Suppressor Cells/pathology , Organ Specificity
10.
J Immunother Cancer ; 8(2)2020 08.
Article in English | MEDLINE | ID: mdl-32843493

ABSTRACT

In recent years, cell therapy technologies have resulted in impressive results in hematologic malignancies. Treatment of solid tumors with chimeric antigen receptor T-cells (CAR-T) has been less successful. Solid tumors present challenges not encountered with hematologic cancers, including high intra-tumoral pressure and ineffective CAR-T trafficking to the site of disease. Novel delivery methods may enable CAR-T therapies for solid tumor malignancies. A patient with liver metastases secondary to pancreatic adenocarcinoma received CAR-T targeting carcinoembryonic antigen (CEA). Previously we reported that Pressure-Enabled Drug Delivery (PEDD) enhanced CAR-T delivery to liver metastases 5.2-fold. Three doses of anti-CEA CAR-T were regionally delivered via hepatic artery infusion (HAI) using PEDD technology to optimize the therapeutic index. Interleukin-2 was systemically delivered by continuous intravenous infusion to support CAR-T in vivo. HAI of anti-CEA CAR-T was not associated with any serious adverse events (SAEs) above grade 3 and there were no on-target/off-tumor SAEs. Following CAR-T treatment, positron emission tomography-CT demonstrated a complete metabolic response within the liver, which was durable and sustained for 13 months. The response was accompanied by normalization of serum tumor markers and an abundance of CAR+ cells found within post-treatment tumor specimens. The findings from this report exhibit biologic activity and safety of regionally infused CAR-T for an indication with limited immune-oncology success to date. Further studies will determine how HAI of CAR-T may be included in multidisciplinary treatment plans for patients with liver metastases. ClinicalTrials.gov number, NCT02850536.


Subject(s)
Liver Neoplasms/genetics , Drug Delivery Systems , Humans , Immunotherapy/methods , Liver Neoplasms/pathology , Male , Middle Aged , Receptors, Chimeric Antigen/metabolism , Tumor Microenvironment
11.
Cancer Gene Ther ; 27(5): 341-355, 2020 05.
Article in English | MEDLINE | ID: mdl-31155611

ABSTRACT

Effective chimeric antigen receptor-modified T-cell (CAR-T) therapy for liver metastases (LM) will require innovative solutions to ensure efficient delivery and minimization of systemic toxicity. We previously demonstrated the safety of CAR-T hepatic artery infusions (HAI). We subsequently conducted the phase 1b HITM-SIR trial, in which six patients (pts) with CEA+ LM received anti-CEA CAR-T HAIs and selective internal radiation therapy (SIRT). The primary endpoint was safety with secondary assessments of biologic activity. Enrolled pts had a mean LM size of 6.4 cm, 4 pts had >10 LM, and pts received an average of two lines of prior systemic therapy. No grade 4 or 5 toxicities were observed, and there were no instances of severe cytokine-release syndrome (CRS) or neurotoxicity. The mean transduction efficiency was 60.4%. Following CAR-T HAI, reduced levels of GM-CSF-R, IDO, and PD-L1 were detected in LM, and serum CEA levels were stable or decreased in all subjects. Median survival time was 8 months (mean 11, range 4-31). Anti-CEA CAR-T HAI with subsequent SIRT was well tolerated, and biologic responses were demonstrated following failure of conventional therapy. HAI of CAR-T was once again confirmed not to be associated with severe CRS or neurotoxicity.


Subject(s)
Brachytherapy/methods , Immunotherapy, Adoptive/methods , Liver Neoplasms/therapy , Adult , Aged , Carcinoembryonic Antigen/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Combined Modality Therapy/methods , Female , Follow-Up Studies , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/metabolism , Humans , Infusions, Intra-Arterial , Liver/blood supply , Liver/immunology , Liver/pathology , Liver/radiation effects , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Middle Aged , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Rectal Neoplasms/immunology , Rectal Neoplasms/mortality , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Response Evaluation Criteria in Solid Tumors
12.
Oncogene ; 38(4): 533-548, 2019 01.
Article in English | MEDLINE | ID: mdl-30158673

ABSTRACT

Immunosuppressive myeloid-derived suppressor cells (MDSC) subvert antitumor immunity and limit the efficacy of chimeric antigen receptor T cells (CAR-T). Previously, we reported that the GM-CSF/JAK2/STAT3 axis drives liver-associated MDSC (L-MDSC) proliferation and blockade of this axis rescued antitumor immunity. We extended these findings in our murine liver metastasis (LM) model, by treating tumor-bearing mice with STAT3 inhibitors (STATTIC or BBI608) to further our understanding of how STAT3 drives L-MDSC suppressive function. STAT3 inhibition caused significant reduction of tumor burden as well as L-MDSC frequencies due to decrease in pSTAT3 levels. L-MDSC isolated from STATTIC or BBI608-treated mice had significantly reduced suppressive function. STAT3 inhibition of L-MDSC was associated with enhanced antitumor activity of CAR-T. Further investigation demonstrated activation of apoptotic signaling pathways in L-MDSC following STAT3 inhibition as evidenced by an upregulation of the pro-apoptotic proteins Bax, cleaved caspase-3, and downregulation of the anti-apoptotic protein Bcl-2. Accordingly, there was also a decrease of pro-survival markers, pErk and pAkt, and an increase in pro-death marker, Fas, with activation of downstream JNK and p38 MAPK. These findings represent a previously unrecognized link between STAT3 inhibition and Fas-induced apoptosis of MDSCs. Our findings suggest that inhibiting STAT3 has potential clinical application for enhancing the efficacy of CAR-T cells in LM through modulation of L-MDSC.


Subject(s)
Adenocarcinoma/secondary , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Benzofurans/therapeutic use , Cyclic S-Oxides/therapeutic use , Liver Neoplasms, Experimental/secondary , Molecular Targeted Therapy , Myeloid-Derived Suppressor Cells/pathology , Naphthoquinones/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , bcl-2-Associated X Protein/physiology , fas Receptor/physiology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Benzofurans/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cyclic S-Oxides/pharmacology , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , Immunotherapy , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthoquinones/pharmacology , Neoplasm Proteins/physiology , STAT3 Transcription Factor/physiology , Signal Transduction , Specific Pathogen-Free Organisms , Tumor Burden , Tumor Escape/physiology , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics
14.
J Leukoc Biol ; 102(2): 201-208, 2017 08.
Article in English | MEDLINE | ID: mdl-28546503

ABSTRACT

Chimeric antigen receptor expressing T cells (CAR-T) are a promising form of immunotherapy, but the influence of age-related immune changes on CAR-T production remains poorly understood. We showed that CAR-T cells from geriatric donors (gCAR-T) are functionally impaired relative to CAR-T from younger donors (yCAR-T). Higher transduction efficiencies and improved cell expansion were observed in yCAR-T cells compared with gCAR-T. yCAR-T demonstrated significantly increased levels of proliferation and signaling activation of phosphorylated (p)Erk, pAkt, pStat3, and pStat5. Furthermore, yCAR-T contained higher proportions of CD4 and CD8 effector memory (EM) cells, which are known to have enhanced cytolytic capabilities. Accordingly, yCAR-T demonstrated higher levels of tumor antigen-specific cytotoxicity compared with gCAR-T. Enhanced tumor killing by yCAR-T correlated with increased levels of perforin and granzyme B. yCAR-T had increased α5ß1 integrin expression, a known mediator of retroviral transduction. We found that treatment with M-CSF or TGF-ß1 rescued the impaired transduction efficiency of the gCAR-T by increasing the α5ß1 integrin expression. Neutralization of α5ß1 confirmed that this integrin was indispensable for CAR expression. Our study suggests that the increase of α5ß1 integrin expression levels enhances CAR expression and thereby improves tumor killing by gCAR-T.


Subject(s)
Aging/immunology , Immunotherapy/methods , Integrin alpha5beta1/biosynthesis , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Adult , Aged , Blotting, Western , Chimera , Cytotoxicity, Immunologic/immunology , Female , Flow Cytometry , Humans , Integrin alpha5beta1/immunology , Lymphocyte Activation/immunology , Male , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Young Adult
15.
Proc Natl Acad Sci U S A ; 113(9): 2472-7, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26884170

ABSTRACT

Ligands of the endothelial-enriched tunica interna endothelial cell kinase 2 (Tie2) are markedly imbalanced in severe infections associated with vascular leakage, yet regulation of the receptor itself has been understudied in this context. Here, we show that TIE2 gene expression may constitute a novel vascular barrier control mechanism in diverse infections. Tie2 expression declined rapidly in wide-ranging models of leak-associated infections, including anthrax, influenza, malaria, and sepsis. Forced Tie2 suppression sufficed to attenuate barrier function and sensitize endothelium to permeability mediators. Rapid reduction of pulmonary Tie2 in otherwise healthy animals attenuated downstream kinase signaling to the barrier effector vascular endothelial (VE)-cadherin and induced vascular leakage. Compared with wild-type littermates, mice possessing one allele of Tie2 suffered more severe vascular leakage and higher mortality in two different sepsis models. Common genetic variants that influence TIE2 expression were then sought in the HapMap3 cohort. Remarkably, each of the three strongest predicted cis-acting SNPs in HapMap3 was also associated with the risk of acute respiratory distress syndrome (ARDS) in an intensive care unit cohort of 1,614 subjects. The haplotype associated with the highest TIE2 expression conferred a 28% reduction in the risk of ARDS independent of other major clinical variables, including disease severity. In contrast, the most common haplotype was associated with both the lowest TIE2 expression and 31% higher ARDS risk. Together, the results implicate common genetic variation at the TIE2 locus as a determinant of vascular leak-related clinical outcomes from common infections, suggesting new tools to identify individuals at unusual risk for deleterious complications of infection.


Subject(s)
Capillary Permeability , Infections/physiopathology , Receptor, TIE-2/genetics , Animals , Endothelium, Vascular/physiopathology , Mice
16.
Cancer Immunol Immunother ; 64(7): 817-29, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25850344

ABSTRACT

Chimeric antigen receptor-modified T cell (CAR-T) technology, a promising immunotherapeutic tool, has not been applied specifically to treat liver metastases (LM). While CAR-T delivery to LM can be optimized by regional intrahepatic infusion, we propose that liver CD11b+Gr-1+ myeloid-derived suppressor cells (L-MDSC) will inhibit the efficacy of CAR-T in the intrahepatic space. We studied anti-CEA CAR-T in a murine model of CEA+ LM and identified mechanisms through which L-MDSC expand and inhibit CAR-T function. We established CEA+ LM in mice and studied purified L-MDSC and responses to treatment with intrahepatic anti-CEA CAR-T infusions. L-MDSC expanded threefold in response to LM, and their expansion was dependent on GM-CSF, which was produced by tumor cells. L-MDSC utilized PD-L1 to suppress anti-tumor responses through engagement of PD-1 on CAR-T. GM-CSF, in cooperation with STAT3, promoted L-MDSC PD-L1 expression. CAR-T efficacy was rescued when mice received CAR-T in combination with MDSC depletion, GM-CSF neutralization to prevent MDSC expansion, or PD-L1 blockade. As L-MDSC suppressed anti-CEA CAR-T, infusion of anti-CEA CAR-T in tandem with agents targeting L-MDSC is a rational strategy for future clinical trials.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoembryonic Antigen/immunology , Liver Neoplasms/pathology , Myeloid Cells/immunology , Recombinant Fusion Proteins/therapeutic use , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Liver/cytology , Liver/pathology , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , STAT3 Transcription Factor/metabolism , Tumor Burden
17.
Methods Mol Biol ; 1172: 201-8, 2014.
Article in English | MEDLINE | ID: mdl-24908307

ABSTRACT

Immunohistochemical (IHC) staining is an invaluable, sensitive, and effective method to detect the presence and localization of proteins in the cellular compartment in tissues. The basic concept of IHC is detecting the antigen in tissues by means of specific antibody binding, which is then demonstrated with a colored histochemical reaction that can be observed under a light microscope. The most challenging aspect of IHC techniques is optimizing the precise experimental conditions that are required to get a specific and a strong signal. The critical steps of IHC are specimen acquisition, fixation, permeabilization, detection system, and selection of the antigen specific antibody and its optimization. Here, we elaborate the technique using the endothelial growth factor binding receptor Tie2 in mouse lungs.


Subject(s)
Immunohistochemistry/methods , Receptor, TIE-2/metabolism , 3,3'-Diaminobenzidine/chemistry , Animals , Antibodies/chemistry , Avidin/chemistry , Biotin/chemistry , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression , Hematoxylin , Horseradish Peroxidase/chemistry , Hydrogen Peroxide/chemistry , Lung/cytology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Receptor, TIE-2/analysis , Receptor, TIE-2/genetics
18.
Cell Stem Cell ; 12(4): 407-12, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23352605

ABSTRACT

The prospects for using autologous induced pluripotent stem cells (iPSCs) in cell replacement therapy have been tempered by evidence that undifferentiated, syngeneic mouse iPSCs are immunogenic upon transplantation. However, the immunogenicity of more therapeutically relevant differentiated cells remains unexplored. Here, we differentiated mouse iPSCs into embryoid bodies (EBs) or representative cell types spanning the three embryonic germ layers and assessed their immunogenicity in vitro and after their transplantation into syngeneic recipients. We found no evidence of increased T cell proliferation in vitro, rejection of syngeneic iPSC-derived EBs/tissue-specific cells (TSCs) after transplantation, or an antigen-specific secondary immune response. Thus, differentiated cells derived from syngeneic iPSCs do not appear to be rejected after transplantation. We also found little evidence of an immune response to undifferentiated, syngeneic iPSCs. Our data support the idea that differentiated cells generated from autologous iPSCs could be applied for cell replacement therapy without eliciting immune rejection.


Subject(s)
Cell Differentiation/immunology , Immunity/immunology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Graft Rejection/immunology , Graft Survival/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transplantation, Isogeneic
19.
J Clin Invest ; 121(3): 1026-43, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21339643

ABSTRACT

LEOPARD syndrome (LS) is an autosomal dominant "RASopathy" that manifests with congenital heart disease. Nearly all cases of LS are caused by catalytically inactivating mutations in the protein tyrosine phosphatase (PTP), non-receptor type 11 (PTPN11) gene that encodes the SH2 domain-containing PTP-2 (SHP2). RASopathies typically affect components of the RAS/MAPK pathway, yet it remains unclear how PTPN11 mutations alter cellular signaling to produce LS phenotypes. We therefore generated knockin mice harboring the Ptpn11 mutation Y279C, one of the most common LS alleles. Ptpn11(Y279C/+) (LS/+) mice recapitulated the human disorder, with short stature, craniofacial dysmorphia, and morphologic, histologic, echocardiographic, and molecular evidence of hypertrophic cardiomyopathy (HCM). Heart and/or cardiomyocyte lysates from LS/+ mice showed enhanced binding of Shp2 to Irs1, decreased Shp2 catalytic activity, and abrogated agonist-evoked Erk/Mapk signaling. LS/+ mice also exhibited increased basal and agonist-induced Akt and mTor activity. The cardiac defects in LS/+ mice were completely reversed by treatment with rapamycin, an inhibitor of mTOR. Our results demonstrate that LS mutations have dominant-negative effects in vivo, identify enhanced mTOR activity as critical for causing LS-associated HCM, and suggest that TOR inhibitors be considered for treatment of HCM in LS patients.


Subject(s)
Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/genetics , Immunosuppressive Agents/pharmacology , LEOPARD Syndrome/drug therapy , LEOPARD Syndrome/genetics , Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Sirolimus/pharmacology , Animals , Catalysis , Echocardiography , Female , Humans , Male , Mice , Phenotype , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
20.
Arch Biochem Biophys ; 490(2): 85-95, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19682972

ABSTRACT

We have recently identified two promoters, distal and proximal for rat mitochondrial glycerophosphate acyltransferase (mtGPAT). Here we are reporting further characterization of the promoters. Insulin and epidermal growth factor (EGF) stimulated while leptin and glucagon inhibited the luciferase activity of the distal promoter and the amounts of the distal transcript. Conversely, luciferase activity of the proximal promoter and proximal transcript remained unchanged due to these treatments. Only the distal promoter has binding sites for carbohydrate response element binding protein (ChREBP) and sterol regulatory element binding protein-1 (SREBP-1). Electromobility shift assays and chromatin immunoprecipitation assays demonstrated that ChREBP and SREBP-1 bind to the mtGPAT distal promoter. Insulin and EGF increased while glucagon and leptin decreased the binding of SREBP-1 and ChREBP to the distal promoter. Thus, the distal promoter is the regulatory promoter while the proximal promoter acts constitutively for rat mtGPAT gene under the influence of hormones and growth factor.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glycerol-3-Phosphate O-Acyltransferase/genetics , Mitochondria, Liver/enzymology , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA Primers/genetics , Epidermal Growth Factor/pharmacology , Glucagon/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin/pharmacology , Leptin/pharmacology , Luciferases/genetics , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , Rats , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL