Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
1.
J Transl Med ; 22(1): 220, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38429732

ABSTRACT

BACKGROUND: Targeting CD47/SIRPα axis has emerged as a promising strategy in cancer immunotherapy. Despite the encouraging clinical efficacy observed in hematologic malignancies through CD47-SIRPα blockade, there are safety concerns related to the binding of anti-CD47 antibodies to CD47 on the membrane of peripheral blood cells. METHODS: In order to enhance the selectivity and therapeutic efficacy of the antibody, we developed a humanized anti-CD47 monoclonal antibody called Gentulizumab (GenSci059). The binding capacity of GenSci059 to CD47 was evaluated using flow cytometry and surface plasmon resonance (SPR) methods, the inhibitory effect of GenSci059 on the CD47-SIRPα interaction was evaluated through competitive ELISA assays. The anti-tumor activity of GenSci059 was assessed using in vitro macrophage models and in vivo patient-derived xenograft (PDX) models. To evaluate the safety profile of GenSci059, binding assays were conducted using blood cells. Additionally, we investigated the underlying mechanisms contributing to the weaker binding of GenSci059 to erythrocytes. Finally, toxicity studies were performed in non-human primates to assess the potential risks associated with GenSci059. RESULTS: GenSci059 displayed strong binding to CD47 in both human and monkey, and effectively inhibited the CD47-SIRPα interaction. With doses ranging from 5 to 20 mg/kg, GenSci059 demonstrated potent inhibition of the growth of subcutaneous tumor with the inhibition rates ranged from 30.3% to complete regression. Combination of GenSci059 with 2.5 mg/kg Rituximab at a dose of 2.5 mg/kg showed enhanced tumor inhibition compared to monotherapy, exhibiting synergistic effects. GenSci059 exhibited minimal binding to hRBCs compared to Hu5F9-G4. The binding of GenSci059 to CD47 depended on the cyclization of N-terminal pyroglutamic acid and the spatial conformation of CD47, but was not affected by its glycosylation modifications. A maximum tolerated dose (MTD) of 450 mg/kg was observed for GenSci059, and no significant adverse effects were observed in repeated dosages up to 10 + 300 mg/kg, indicating a favorable safety profile. CONCLUSION: GenSci059 selectively binds to CD47, effectively blocks the CD47/SIRPα axis signaling pathway and enhances the phagocytosis effects of macrophages toward tumor cells. This monoclonal antibody demonstrates potent antitumor activity and exhibits a favorable safety profile, positioning it as a promising and effective therapeutic option for cancer.


Subject(s)
CD47 Antigen , Neoplasms , Animals , Humans , Neoplasms/pathology , Phagocytosis , Macrophages/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Disease Models, Animal , Antigens, Differentiation/metabolism , Antigens, Differentiation/pharmacology , Antigens, Differentiation/therapeutic use
2.
BMC Complement Med Ther ; 24(1): 10, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167059

ABSTRACT

BACKGROUND: Shikonin, a natural naphthoquinone compound extracted from the Chinese traditional herbal medicine "Lithospermum erythrorhizon", possesses antitumor activity against various cancer types. Tumor-suppressor genes (TSGs) negatively regulate cell growth, proliferation, and differentiation, thereby inhibiting tumor formation. However, the molecular mechanism of action of shikonin on TSGs in non-small-cell lung cancer (NSCLC) remains unclear. METHODS: The inhibitory effect of shikonin on the proliferation and migration abilities of lung cancer cells were measured by Cell Counting Kit 8 (CCK8) and wound healing assays. The alteration of genes by shikonin treatment was detected by mRNA high-throughput sequencing and further confirmed by qPCR and western blotting experiments. The dominant functions of the upregulated genes were analyzed by GO and KEGG profiling. RESULTS: Shikonin inhibited the proliferation and migration of A549 and H1299 NSCLC cells in a dose-dependent manner. mRNA high-throughput sequencing revealed a total of 1794 upregulated genes in shikonin-treated NSCLC cells. Moreover, bioinformatic analysis of GO and KEGG profiling revealed that the up-regulated genes were mostly involved in the JNK/P38/MAPK signaling pathway, among which the expression of GADD45B and PPP3CC was significantly enhanced. Finally, we confirmed that GADD45B and PPP3CC were indeed upregulated in JNK/P38/MAPK pathway. CONCLUSIONS: Taken together, these results suggested that shikonin might affect the expression of GADD45B and PPP3CC through the JNK/P38/MAPK pathway, therefore exerting an inhibitory effect on the proliferation and migration of cancer cells. To our knowledge, this is the first study reporting the role of shikonin in upregulating TSGs to activate the JNK/P38/MAPK signaling pathways in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Naphthoquinones , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Cell Line, Tumor , MAP Kinase Signaling System , Naphthoquinones/pharmacology , Cell Proliferation , RNA, Messenger/metabolism , GADD45 Proteins , Antigens, Differentiation/metabolism , Antigens, Differentiation/pharmacology , Calcineurin/metabolism , Calcineurin/pharmacology
3.
Anim Biotechnol ; 35(1): 2298399, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38157229

ABSTRACT

Cholesterol is regarded as a signaling molecule in regulating the metabolism and function of fat cells, in which 7-Dehydrocholesterol reductase (DHCR7) is a key enzyme that catalyzes the conversion of 7-dehydrocholesterol to cholesterol, however, the exact function of DHCR7 in goat adipocytes remains unknown. Here, the effect of DHCR7 on the formation of subcutaneous and intramuscular fat in goats was investigated in vitro, and the result indicated that the mRNA level of DHCR7 showed a gradual downward trend in subcutaneous adipogenesis, but an opposite trend in intramuscular adipogenesis. In the process of subcutaneous preadipocytes differentiation, overexpression of DHCR7 inhibited the expression of adipocytes differentiation marker genes (CEBP/α, CEBP/ß, SREBP1 and AP2), lipid metabolism-related genes (AGPAT6, FASN, SCD1 and LPL), and the lipid accumulation. However, in intramuscular preadipocyte differentiation, DHCR7 overexpression showed a promoting effect on adipocyte differentiation marker genes (CEBP/α, CEBP/ß, PPARγ and SREBP1) and lipid metabolism-related genes (GPAM, AGPAT6, DGAT1 and SCD1) expression, and on lipid accumulation. In summary, our work demonstrated that DHCR7 played an important role in regulating adipogenic differentiation and lipid metabolism in preadipocytes in goats, which is of great significance for uncovering the underlying molecular mechanism of adipocyte differentiation and improving goat meat quality.


Subject(s)
Goats , Oxidoreductases , Animals , Goats/genetics , Cell Differentiation/genetics , Adipogenesis/genetics , Adipocytes/metabolism , Antigens, Differentiation/metabolism , Antigens, Differentiation/pharmacology , Cholesterol/metabolism , Lipids , PPAR gamma/metabolism
4.
Cancer Immunol Immunother ; 72(9): 2879-2888, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37217603

ABSTRACT

The use of treatments, such as programmed death protein 1 (PD1) or cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies, that loosen the natural checks upon immune cell activity to enhance cancer killing have shifted clinical practice and outcomes for the better. Accordingly, the number of antibodies and engineered proteins that interact with the ligand-receptor components of immune checkpoints continue to increase along with their use. It is tempting to view these molecular pathways simply from an immune inhibitory perspective. But this should be resisted. Checkpoint molecules can have other cardinal functions relevant to the development and use of blocking moieties. Cell receptor CD47 is an example of this. CD47 is found on the surface of all human cells. Within the checkpoint paradigm, non-immune cell CD47 signals through immune cell surface signal regulatory protein alpha (SIRPα) to limit the activity of the latter, the so-called trans signal. Even so, CD47 interacts with other cell surface and soluble molecules to regulate biogas and redox signaling, mitochondria and metabolism, self-renewal factors and multipotency, and blood flow. Further, the pedigree of checkpoint CD47 is more intricate than supposed. High-affinity interaction with soluble thrombospondin-1 (TSP1) and low-affinity interaction with same-cell SIRPα, the so-called cis signal, and non-SIRPα ectodomains on the cell membrane suggests that multiple immune checkpoints converge at and through CD47. Appreciation of this may provide latitude for pathway-specific targeting and intelligent therapeutic effect.


Subject(s)
CD47 Antigen , Neoplasms , Humans , Antigens, Differentiation/pharmacology , Receptors, Immunologic/metabolism , Neoplasms/therapy , Antibodies/pharmacology , Carrier Proteins , Phagocytosis
5.
Cell Mol Biol (Noisy-le-grand) ; 69(14): 40-44, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38279488

ABSTRACT

This study aimed to explore the correlation between the expression of phosphatase and tensin homolog (PTEN), a tumor suppressor gene, and CD47-SIRPα signaling pathway, and clarify the underlying mechanisms of the bone tumor inhibition effect of PTEN. In this study, August x Copenhagen Irish (ACI) male rats were used, and 100µl of UMR-106 cell suspension (1´106 cells) was injected subcutaneously to induce the bone tumor model. The gene expression of PTEN, CD47 and SIRPα of both groups (control and bone tumor model) were analyzed by RT-PCR. For in vitro experiments, pEGFP-N1-PTEN plasmid was used to transfect the murine bone tumor UMR-106 and the human bone tumor KRIB cells. Also, we detected the invasiveness of the UMR-106 cells and KRIB cells after transfection. In this study, we observed that the gene expression of PTEN and SIRPα were significantly decreased in the ACI rats with bone tumors in comparison to the control group, however, the expression level of CD47 has been significantly increased. The tumor cells transfected with the pEGFP-N1-PTEN plasmid showed significantly higher levels of PTEN expression, however, the expression level of the CD47 gene has been decreased. Also, the invasion ability of tumor cells has been down-regulated. Also, we observed a negative correlation between the gene expression of the tumor suppressor gene PTEN and the CD47 and SIRPα genes. In summary, based on the anti-tumor effect of PTEN and its effect on inhibition of the bone tumor, it could be hypothesized that this phenomenon might be related to the phosphorylation of the CD47 and SIRPα gene.


Subject(s)
Bone Neoplasms , CD47 Antigen , Animals , Humans , Male , Mice , Rats , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Antigens, Differentiation/pharmacology , Bone Neoplasms/genetics , CD47 Antigen/genetics , CD47 Antigen/metabolism , Phagocytosis , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Receptors, Immunologic/genetics , Signal Transduction
6.
Clin Transl Med ; 12(8): e943, 2022 08.
Article in English | MEDLINE | ID: mdl-35908284

ABSTRACT

BACKGROUND: The CD47-signal regulatory protein alpha (SIRPα) 'don't eat me' signalling axis is perhaps the most prominent innate immune checkpoint to date. However, from initial clinical trials, it is evident that monotherapy with CD47-SIRPα blocking has a limited therapeutic effect at the maximum tolerated dose. Furthermore, treatment is associated with severe side effects, most notably anaemia, that are attributable to the ubiquitous expression of CD47. Nevertheless, promising clinical responses have been reported upon combination with the tumour-targeting antibody rituximab or azacytidine, although toxicity issues still hamper clinical application. MAIN BODY: Here, we discuss the current state of CD47-SIRPα blocking therapy with a focus on limitations of current strategies, such as depletion of red blood cells. Subsequently, we focus on innovations designed to overcome these limitations. These include novel antibody formats designed to selectively target CD47 on tumour cells as well as tumour-targeted bispecific antibodies with improved selectivity. In addition, the rationale and outcome of combinatorial approaches to improve the therapeutic effect of CD47 blockade are discussed. Such combinations include those with tumour-targeted opsonizing antibodies, systemic therapy, epigenetic drugs, other immunomodulatory T-cell-targeted therapeutics or dual immunomodulatory CD47 bispecific antibodies. CONCLUSION: With these advances in the design of CD47-SIRPα-targeting therapeutic strategies and increasing insight into the mechanism of action of this innate checkpoint, including the role of adaptive immunity, further advances in the clinical application of this checkpoint can be anticipated.


Subject(s)
Antibodies, Bispecific , Antigens, Differentiation/metabolism , Neoplasms , Receptors, Immunologic/metabolism , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antigens, Differentiation/pharmacology , CD47 Antigen/metabolism , CD47 Antigen/therapeutic use , Humans , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunotherapy , Neoplasms/drug therapy , Phagocytosis , Receptors, Immunologic/therapeutic use
7.
Colloids Surf B Biointerfaces ; 217: 112609, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35667200

ABSTRACT

Acute myeloid leukemia (AML), a malignant disorder of Hematopoietic stem cells, can escape immunosurveillance by over expression of the cluster of differentiation 47 (CD47) marker, which functions as an inhibitory signal, suppressing phagocytosis by binding to signal regulatory protein α (SIRPα) on macrophages. AML is treated mainly by chemotherapy, which has drastic side effects and poor outcomes for the patients. Most AML patients develop drug resistance, so other methods to treat AML are highly required. Small interfering RNA (siRNA) is considered as an antitumor therapeutic due to its ability to silence genes associated with the overexpressed cancer markers and subsequently re-sensitize cancer cells. However, delivering siRNA into cells faces challenges, and the development of an effective delivery system is desired for successful silencing at the gene level. Herein, we report the usage of different formulations of graphene oxide (GO) as carriers for the delivery of CD47_siRNA (siRNA against CD47) into AML cells in vitro. The polyethylene glycol (PEG) and dendrimers (PAMAM) modified GO with small flake sizes achieved the highest silencing efficiency of the anti-phagocytosis marker CD47 gene, resulted CD47 protein down-regulation in AML cells. Moreover, the concentration at which the GO-based formulations was used has shown no cytotoxicity in AML cells or normal blood cells, which could be used to screen potential drugs for targeted gene therapy in AML.


Subject(s)
CD47 Antigen , Leukemia, Myeloid, Acute , Antigens, CD , Antigens, Differentiation/pharmacology , Antigens, Differentiation/therapeutic use , CD47 Antigen/genetics , CD47 Antigen/metabolism , CD47 Antigen/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Phagocytosis , RNA, Small Interfering
8.
Mol Pharm ; 19(5): 1273-1293, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35436123

ABSTRACT

Cancer is still a major disease that is currently difficult for humans to overcome. When the expression of the cluster of differentiation 47 (CD47) is upregulated, tumor cells interact with the macrophage inhibitory receptor signal regulatory protein α (SIRPα) to transmit the "Don't eat me" signal, thereby avoiding phagocytosis by the macrophages. Therefore, when the CD47-SIRPα axis is inhibited, the macrophages' phagocytic function can be restored and can also exert antitumor effects. This Review mainly introduces recent advances in tumor therapy targeted on the CD47-SIRPα axis, including the antibody and fusion protein, small molecule, gene therapy, cell therapy, and drug delivery system, to inhibit the function of CD47 expressed on tumor cells and promote tumor phagocytosis by macrophages. In addition, this Review also summarizes the current approaches to avoid anemia, a common side effect of CD47-SIRPα inhibitions, and provides ideas for clinical transformation.


Subject(s)
CD47 Antigen , Neoplasms , Antigens, Differentiation/metabolism , Antigens, Differentiation/pharmacology , Humans , Immunotherapy , Neoplasms/drug therapy , Phagocytosis , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
9.
Cancer Immunol Immunother ; 70(11): 3291-3302, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33852044

ABSTRACT

Cancer immunotherapies, including immune checkpoint blockage and adoptive transfer of CAR-T cells, have achieved historical successes for many kinds of malignancy. However, a minority of patients survive long term over 5 years without relapse, perhaps owing to tumor heterogeneity and potent immunosuppression in the tumor microenvironment. Here, using an established mouse tumor model of triple-negative 4T1 breast cancer, we show that local immunochemotherapy triggers powerful local and systemic antitumor immunity. Paraneoplastic injection of CpG, α-OX40, and anthracycline completely eliminated both local and distant large established 4T1 breast cancer without obvious relapse. Analysis of the immune cells at tumor tissues, draining lymph nodes, and spleens revealed that the local treatment increased the infiltration of CD4+ and CD8+ T cells in all three tissues and inhibited the accumulation of myeloid-derived suppressor cells in the spleen in a delayed response. Most importantly, this treatment triggered systemic T cell response against 4T1 tumors and some of their neoantigen epitopes as detected by IFN-γ ELISpot and intracellular cytokine assays in splenocytes. Furthermore, T cells showed specific cytotoxic activity against 4T1 tumor cells in vitro. In general, this local immunochemotherapy provides a new approach to target highly diverse neoantigens in various types of cancers without complicated and expensive antigen identification via next-generation sequencing.


Subject(s)
Antigens, Differentiation/pharmacology , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Oligodeoxyribonucleotides/pharmacology , T-Lymphocytes/immunology , Triple Negative Breast Neoplasms/immunology , Animals , Female , Immunotherapy , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
Cancer Immunol Immunother ; 69(12): 2561-2569, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32583154

ABSTRACT

Many cancer cells express CD47 as a 'don't eat me' signal to mask their presences from immune recognition and destruction. Such a signal is transmitted when CD47 binds to the signal regulatory protein-α (SIRPα) on macrophages to cut the phagocytic reaction. Most recent studies have focused on developing CD47 blocking agents with different affinities and avidities in order to optimize the therapeutic window between efficacy and toxicities involving normal cells expressing CD47. We described in this study a new design to fuse one CD47 binding domain of SIRPα with a pharmacokinetics modifying domain F8. The resulted single valent long-acting CD47 antagonist SIRPα-F8 was able to bind to CD47 and disrupt CD47-SIRPα axis. However, by itself it cannot trigger endocytosis and has no effect on tumor growth. Only when used in combination with the anti-CD20 mAbs, there were greatly improved phagocytic activities towards CD20 positive cancer cells. In vivo the combination also resulted in better tumor growth inhibition comparing to the vehicle control group. In addition, we showed that the F8 fusion bound to hFcRn only inside endosomes at pH 6.0, enabled hFcRn mediated recycling and thus greatly extended the circulation half-life in hFcRn knock-in mice. Taken together, the SIRPα-F8 design may suggest a new option to improve the therapeutic index of antibody treatment in clinical use towards tumors.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CD47 Antigen/antagonists & inhibitors , Neoplasms/drug therapy , Phagocytosis/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Differentiation/genetics , Antigens, Differentiation/pharmacology , Antigens, Differentiation/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/immunology , CHO Cells , Cell Line, Tumor , Cricetulus , Gene Knock-In Techniques , HEK293 Cells , Histocompatibility Antigens Class I/genetics , Humans , Injections, Intralesional , Macrophages , Mice, Transgenic , Monocytes , Neoplasms/immunology , Neoplasms/pathology , Phagocytosis/immunology , Receptors, Fc/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/therapeutic use , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Xenograft Model Antitumor Assays
11.
Viruses ; 12(3)2020 03 08.
Article in English | MEDLINE | ID: mdl-32182730

ABSTRACT

Interferon-inducible transmembrane proteins (IFITM1/2/3) have been reported to suppress the entry of a wide range of viruses. However, their antiviral functional residues and specific mechanisms are still unclear. Here, we firstly resolved the topology of IFITM1 on the plasma membrane where N-terminus points into the cytoplasm and C-terminus resides extracellularly. Further, KRRK basic residues of IFITM1 locating at 62-67 of the conserved intracellular loop (CIL) were found to play a key role in the restriction on the Zika virus (ZIKV) and dengue virus (DENV). Similarly, KRRK basic residues of IFITM2/3 also contributed to suppressing ZIKV replication. Finally, IFITM1 was revealed to be capable of restricting the release of ZIKV particles from endosome to cytosol so as to impede the entry of ZIKV into host cells, which was tightly related with the inhibition of IFITM1 on the acidification of organelles. Overall, our study provided topology, antiviral functional residues and the mechanism of interferon-inducible transmembrane proteins.


Subject(s)
Antigens, Differentiation/metabolism , Antigens, Differentiation/pharmacology , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Zika Virus/drug effects , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Chlorocebus aethiops , Endosomes/metabolism , HEK293 Cells , Humans , Interferons/metabolism , Membrane Proteins/metabolism , Sequence Alignment , Vero Cells , Virus Replication/drug effects , Zika Virus Infection
12.
Int J Biol Macromol ; 151: 1181-1193, 2020 May 15.
Article in English | MEDLINE | ID: mdl-31743714

ABSTRACT

Interferon-inducible transmembrane proteins (IFITMs) restrict infection by several viruses, such as influenza A virus, West Nile virus and dengue virus. It has not been determined whether porcine IFITMs (pIFITMs) inhibit infection by pseudorabies virus (PRV), an enveloped, double-stranded DNA virus, which is the etiological agent of Aujeszky's disease in pigs. Here, we report that PRV infection elicited pIFITM1 expression in PK15 porcine kidney epithelial cells and 3D4/21 alveolar macrophages. pIFITM2 and pIFITM3 expression was only elevated in PK15 cells during PRV infection. Depletion of pIFITM1 using RNA interference, either in PK15 or in 3D4/21 cells, enhanced PRV infection while overexpression of pIFITM1 had the opposite effect. Knockdown of pIFITM2 and pIFITM3 did not influence PRV infection, suggesting that pIFITM2 and pIFITM3 are independent of PRV infection. PRV-induced pIFITM1 expression was dependent on the cGAS/STING/TBK1/IRF3 innate immune pathway and interferon-alpha receptor-1, suggesting that pIFITM1 is up-regulated by the type I interferon signaling pathway. The anti-PRV role of pIFITM1 was inhibited upon PRV entry. Our data demonstrate that pIFITM1 is a host restriction factor that inhibits PRV entry that may shed light on a strategy for prevention of PRV infection.


Subject(s)
Antigens, Differentiation/pharmacology , Antiviral Agents/pharmacology , Herpesvirus 1, Suid/drug effects , Amino Acid Sequence , Animals , Cell Line , Gene Expression Regulation , Gene Knockdown Techniques , Host-Pathogen Interactions , Humans , Immunity, Innate , Pseudorabies/genetics , Pseudorabies/metabolism , Pseudorabies/virology , Swine , Virus Internalization/drug effects , Virus Replication/drug effects
13.
Clin Cancer Res ; 25(21): 6406-6416, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31371342

ABSTRACT

PURPOSE: OX40 agonist-based combinations are emerging as a novel avenue to improve the effectiveness of cancer immunotherapy. To better guide its clinical development, we characterized the role of the OX40 pathway in tumor-reactive immune cells. We also evaluated combining OX40 agonists with targeted therapy to combat resistance to cancer immunotherapy.Experimental Design: We utilized patient-derived tumor-infiltrating lymphocytes (TILs) and multiple preclinical models to determine the direct effect of anti-OX40 agonistic antibodies on tumor-reactive CD8+ T cells. We also evaluated the antitumor activity of an anti-OX40 antibody plus PI3Kß inhibition in a transgenic murine melanoma model (Braf mutant, PTEN null), which spontaneously develops immunotherapy-resistant melanomas. RESULTS: We observed elevated expression of OX40 in tumor-reactive CD8+ TILs upon encountering tumors; activation of OX40 signaling enhanced their cytotoxic function. OX40 agonist antibody improved the antitumor activity of CD8+ T cells and the generation of tumor-specific T-cell memory in vivo. Furthermore, combining anti-OX40 with GSK2636771, a PI3Kß-selective inhibitor, delayed tumor growth and extended the survival of mice with PTEN-null melanomas. This combination treatment did not increase the number of TILs, but it instead significantly enhanced proliferation of CD8+ TILs and elevated the serum levels of CCL4, CXCL10, and IFNγ, which are mainly produced by memory and/or effector T cells. CONCLUSIONS: These results highlight a critical role of OX40 activation in potentiating the effector function of tumor-reactive CD8+ T cells and suggest further evaluation of OX40 agonist-based combinations in patients with immune-resistant tumors.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , Melanoma/drug therapy , PTEN Phosphohydrolase/genetics , Receptors, OX40/immunology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Heterografts , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Receptors, OX40/antagonists & inhibitors
14.
Cancer Immunol Res ; 6(2): 201-208, 2018 02.
Article in English | MEDLINE | ID: mdl-29305519

ABSTRACT

Although an immune response to tumors may be generated using vaccines, so far, this approach has only shown minimal clinical success. This is attributed to the tendency of cancer to escape immune surveillance via multiple immune suppressive mechanisms. Successful cancer immunotherapy requires targeting these inhibitory mechanisms along with enhancement of antigen-specific immune responses to promote sustained tumor-specific immunity. Here, we evaluated the effect of indoximod, an inhibitor of the immunosuppressive indoleamine-(2,3)-dioxygenase (IDO) pathway, on antitumor efficacy of anti-OX40 agonist in the context of vaccine in the IDO- TC-1 tumor model. We demonstrate that although the addition of anti-OX40 to the vaccine moderately enhances therapeutic efficacy, incorporation of indoximod into this treatment leads to enhanced tumor regression and cure of established tumors in 60% of treated mice. We show that the mechanisms by which the IDO inhibitor leads to this therapeutic potency include (i) an increment of vaccine-induced tumor-infiltrating effector T cells that is facilitated by anti-OX40 and (ii) a decrease of IDO enzyme activity produced by nontumor cells within the tumor microenvironment that results in enhancement of the specificity and the functionality of vaccine-induced effector T cells. Our findings suggest a translatable strategy to enhance the overall efficacy of cancer immunotherapy. Cancer Immunol Res; 6(2); 201-8. ©2018 AACR.


Subject(s)
Antigens, Differentiation/pharmacology , Lung Neoplasms/drug therapy , Tryptophan Oxygenase/antagonists & inhibitors , Tryptophan/analogs & derivatives , Animals , Antigens, Differentiation/immunology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Epitopes, T-Lymphocyte , Female , Humans , Immunotherapy/methods , Lung Neoplasms/immunology , Mice , Mice, Inbred C57BL , Tryptophan/pharmacology , Tryptophan Oxygenase/immunology , Xenograft Model Antitumor Assays
15.
MAbs ; 10(2): 315-324, 2018.
Article in English | MEDLINE | ID: mdl-29182441

ABSTRACT

The host immune system generally serves as a barrier against tumor formation. Programmed death-ligand 1 (PD-L1) is a critical "don't find me" signal to the adaptive immune system, whereas CD47 transmits an anti-phagocytic signal, known as the "don't eat me" signal, to the innate immune system. These and similar immune checkpoints are often overexpressed on human tumors. Thus, dual targeting both innate and adaptive immune checkpoints would likely maximize anti-tumor therapeutic effect and elicit more durable responses. Herein, based on the variable region of atezolizumab and consensus variant 1 (CV1) monomer, we constructed a dual-targeting fusion protein targeting both CD47 and PD-L1 using "Knobs-into-holes" technology, denoted as IAB. It was effective in inducing phagocytosis of tumor cells, stimulating T-cell activation and mediating antibody-dependent cell-mediated cytotoxicity in vitro. No obvious sign of hematological toxicity was observed in mice administered IAB at a dose of 100 mg/kg, and IAB exhibited potent antitumor activity in an immune-competent mouse model of MC38. Additionally, the anti-tumor effect of IAB was impaired by anti-CD8 antibody or clodronate liposomes, which implied that both CD8+ T cells and macrophages were required for the anti-tumor efficacy of IAB and IAB plays an essential role in the engagement of innate and adaptive immune responses. Collectively, these results demonstrate the capacity of an elicited endogenous immune response against tumors and elucidate essential characteristics of synergistic innate and adaptive immune response, and indicate dual blockade of CD47 and PD-L1 by IAB may be a synergistic therapy that activates both innate and adaptive immune response against tumors.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD47 Antigen/antagonists & inhibitors , Immunotherapy/methods , Tumor Escape/drug effects , Adaptive Immunity/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antigens, Differentiation/pharmacology , Humans , Immunity, Innate/drug effects , Immunoglobulin Variable Region , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Phagocytosis/drug effects , Receptors, Immunologic , Recombinant Fusion Proteins/pharmacology
16.
Oncotarget ; 8(7): 11284-11301, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28061465

ABSTRACT

CD47, expressed on a variety of tumor cells, confers immune resistance by delivering an inhibitory "don't eat me" signal to phagocytic cells via its myeloid-specific receptor SIRPα. Recent studies have shown that blocking the CD47-SIRPα axis with CD47-directed antibodies or antibody-derivatives enhances phagocytosis and increases antitumor immune effects. However, CD47 expression on healthy cells creates an antigen sink and potential sites of toxicity, limiting the efficacy of CD47-directed therapies. In this study, we first characterized CD47 expression in Acute Myeloid Leukemia (AML) patients (n = 213) and found that CD47 is highly expressed on both AML bulk and stem cells irrespective of the disease state. Furthermore, to inhibit the CD47-SIRPα signaling pathway at the tumor site, we developed a so-called local inhibitory checkpoint monoclonal antibody (licMAB) by grafting the endogenous SIRPα domain to the N-terminus of the light chain of an antibody targeting CD33, a surface antigen expressed in AML. LicMABs selectively bind CD33-expressing cells even in the presence of a large CD33-negative CD47-positive antigen sink, stimulate phagocytosis of AML cells and eliminate AML cell lines and primary, patient-derived AML cells. Our findings qualify licMABs as a promising therapeutic approach to confine the benefit of disrupting the CD47-SIRPα axis to tumor antigen-expressing cells.


Subject(s)
Antibodies, Blocking/pharmacology , Antigens, Differentiation/pharmacology , Immunotherapy/methods , Leukemia, Myeloid, Acute/immunology , Phagocytosis/drug effects , Sialic Acid Binding Ig-like Lectin 3/immunology , Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , CD47 Antigen/biosynthesis , CD47 Antigen/immunology , Cell Separation , Flow Cytometry , Humans , Microscopy, Confocal , Receptors, Immunologic
17.
PLoS One ; 10(3): e0118794, 2015.
Article in English | MEDLINE | ID: mdl-25738301

ABSTRACT

The interferon-inducible transmembrane (IFITM) proteins inhibit a wide range of viruses. We previously reported the inhibition of human immunodeficiency virus type 1 (HIV-1) strain BH10 by human IFITM1, 2 and 3. It is unknown whether other HIV-1 strains are similarly inhibited by IFITMs and whether there exists viral countermeasure to overcome IFITM inhibition. We report here that the HIV-1 NL4-3 strain (HIV-1NL4-3) is not restricted by IFITM1 and its viral envelope glycoprotein is partly responsible for this insensitivity. However, HIV-1NL4-3 is profoundly inhibited by an IFITM1 mutant, known as Δ(117-125), which is deleted of 9 amino acids at the C-terminus. In contrast to the wild type IFITM1, which does not affect HIV-1 entry, the Δ(117-125) mutant diminishes HIV-1NL4-3 entry by 3-fold. This inhibition correlates with the predominant localization of Δ(117-125) to the plasma membrane where HIV-1 entry occurs. In spite of strong conservation of IFITM1 among most species, mouse IFITM1 is 19 amino acids shorter at its C-terminus as compared to human IFITM1 and, like the human IFITM1 mutant Δ(117-125), mouse IFITM1 also inhibits HIV-1 entry. This is the first report illustrating the role of viral envelope protein in overcoming IFITM1 restriction. The results also demonstrate the importance of the C-terminal region of IFITM1 in modulating the antiviral function through controlling protein subcellular localization.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Antigens, Differentiation/chemistry , Antigens, Differentiation/pharmacology , HIV-1/drug effects , Amino Acid Sequence , Animals , Antigens, Differentiation/genetics , HEK293 Cells , HIV-1/metabolism , HIV-1/physiology , Humans , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Deletion , Species Specificity , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects
18.
Cancer Immunol Res ; 2(4): 307-19, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24764578

ABSTRACT

A major barrier to vaccines in cancer treatment is their failure to activate and maintain a complete cancer-specific CD8(+) effector T-cell repertoire. Low-avidity T cells are more likely to escape clonal deletion in the thymus when compared with high-avidity T cells, and therefore comprise the major population of effector T cells available for activation in patients with cancer. However, low-avidity T cells fail to traffic into the tumor microenvironment and function in eradicating tumor under optimal vaccination conditions as opposed to high-avidity T cells that escape clonal deletion and function in tumor killing. We used high- and low-avidity T-cell receptor transgenic CD8(+) T cells specific for the immunodominant epitope HER2/neu (RNEU420-429) to identify signaling pathways responsible for the inferior activity of the low-avidity T cells. Adoptive transfer of these cells into tumor-bearing vaccinated mice identified the members of apoptosis pathways that are upregulated in low-avidity T cells. The increased expression of proapoptotic proteins by low-avidity T cells promoted their own cell death and also that of other tumor-specific CD8(+) T cells within their local environment. Importantly, we show that this proapoptotic effect can be overcome by using a strong costimulatory signal that prevents the activation-induced cell death and enables the low-avidity T cells to traffic into the tumor and assist in tumor clearance. These findings identify new therapeutic opportunities for activating the most potent anticancer T-cell responses.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/metabolism , Receptor, ErbB-2/metabolism , T-Cell Antigen Receptor Specificity/immunology , Tumor Escape/immunology , Animals , Antigens, Differentiation/pharmacology , Antigens, Neoplasm/immunology , CD24 Antigen/genetics , CD24 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Humans , Immunodominant Epitopes/immunology , Interferon-gamma/biosynthesis , Mice , Mice, Transgenic , Neoplasms/drug therapy , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
19.
Transplant Proc ; 45(6): 2565-8, 2013.
Article in English | MEDLINE | ID: mdl-23953582

ABSTRACT

AIM: The aim of this study was to investigate cytokine messenger RNA (mRNA) expression by peripheral blood mononuclear cells (PBMCs) from renal recipients experiencing acute rejection by blocking OX40-OX40L interactions with recombinant human OX40-Fc fusion protein (rhOX40Fc) in vitro. METHODS: PBMCs were isolated from 20 recipients experiencing acute rejection episodes (rejection group) and 20 recipients with stable graft function (stable group). Levels of Th1 (interferon [IFN]-γ) and Th2 (interleukin [IL]-4) mRNA expressions by PBMCs were measured using real-time reverse transcriptase-polymerase chain reactions. RESULTS: IFN-γ mRNA expression levels were significantly higher in the rejection than the stable group (P < .05). Levels of IL-4 mRNA expression were not significantly different. Among the rejection group, rhOX40Fc reduced significantly the expression of IFN-γ and IL-4 mRNA by anti-CD3-monoclonal antibody stimulated PBMCs (P < .05, and P < .01, respectively). CONCLUSIONS: Blocking of the interaction between OX40 and OX40L in vitro inhibited production of Thl and Th2 type cytokines.


Subject(s)
Antigens, Differentiation/pharmacology , Cytokines/genetics , Graft Rejection/genetics , Immunoglobulin Fc Fragments/pharmacology , Kidney Transplantation/adverse effects , OX40 Ligand/antagonists & inhibitors , RNA, Messenger/metabolism , Receptors, OX40/antagonists & inhibitors , Adult , Allografts , Case-Control Studies , Cells, Cultured , Down-Regulation , Female , Graft Rejection/immunology , Graft Rejection/metabolism , Humans , Interferon-gamma/genetics , Interleukin-4/genetics , Male , Middle Aged , OX40 Ligand/metabolism , Prospective Studies , Receptors, OX40/metabolism , Signal Transduction/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
20.
Protein Expr Purif ; 85(1): 109-16, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22813925

ABSTRACT

Signal regulatory protein (SIRP) α, a transmembrane protein belonging to the immunoglobulin superfamily, is a receptor for CD47. The interaction between SIRPα and CD47 plays an important role in regulating the phagocytosis of leukemia cells and leukemia stem cells (LSCs) by macrophages. Blocking antibodies against CD47 have been shown to promote phagocytosis of LSCs by macrophages. Here, we consider an alternative way to interrupt the interaction between CD47 and SIRPα. We expressed the extracellular domains of the human SIRPα (hSIRP(ext)) and the human CD47 (hCD47(ext)) in Escherichia coli as Trx fusion proteins, and purified them by using affinity chromatography. We show that the purified fusion protein Trx-SIRP(ext) could interact in vitro with Trx-hCD47(ext). Moreover, Trx-SIRP(ext) could effectively bind to Jurkat T-ALL cells, which expressed CD47 at a high level. CD47(ext), on the other hand, bound to human macrophages. In vitro phagocytosis assay showed that these fusion proteins could enhance the phagocytosis of Jurkat cells by macrophage, with Trx-hSIRP(ext) showed a higher efficiency than Trx-CD47(ext). These results indicated that the soluble Trx-hSIRP(ext) and Trx-CD47(ext) polypeptides could be alternative molecules to interrupt CD47-SIRPα interaction between leukemia cells and macrophages, and might be potentially useful for the targeted therapy of leukemia.


Subject(s)
Antigens, Differentiation/genetics , Antigens, Differentiation/pharmacology , CD47 Antigen/genetics , CD47 Antigen/pharmacology , Escherichia coli/genetics , Macrophages/drug effects , Phagocytosis/drug effects , Receptors, Immunologic/genetics , Antigens, Differentiation/chemistry , Antigens, Differentiation/immunology , CD47 Antigen/chemistry , CD47 Antigen/immunology , Humans , Jurkat Cells , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/immunology , Macrophages/immunology , Protein Refolding , Protein Structure, Tertiary , Receptors, Immunologic/chemistry , Receptors, Immunologic/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...