Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 363
Filter
1.
JCI Insight ; 9(3)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38085594

ABSTRACT

Heterologous polyclonal antibodies (pAb) were shown to possess oncolytic properties a century ago with reported clinical responses. More recent preclinical models confirmed pAb efficacy, though their ability to tackle complex target antigens reduces susceptibility to tumor escape. Owing to the recent availability of glyco-humanized pAb (GH-pAb) with acceptable clinical toxicology profile, we revisited use of pAb in oncology and highlighted their therapeutic potential against multiple cancer types. Murine antitumor pAb were generated after repeated immunization of rabbits with murine tumor cell lines from hepatocarcinoma, melanoma, and colorectal cancers. Antitumor pAb recognized and showed cytotoxicity against their targets without cross-reactivity with healthy tissues. In vivo, pAb are effective alone; moreover, these pAb synergize with immune checkpoint inhibitors like anti-PD-L1 in several cancer models. They elicited an antitumor host immune response and prevented metastases. The anticancer activity of pAb was also confirmed in xenografted NMRI nude mice using GH-pAb produced by repeated immunization of pigs with human tumor cell lines. In conclusion, the availability of bioengineered GH-pAb allows for revisiting of passive immunotherapy with oncolytic pAb to fight against solid tumor and cancer metastasis.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Humans , Rabbits , Animals , Mice , Swine , Mice, Nude , Immunization , Melanoma/therapy , Cell Line, Tumor , Antibodies, Neoplasm/pharmacology
2.
Protein Expr Purif ; 189: 105973, 2022 01.
Article in English | MEDLINE | ID: mdl-34560256

ABSTRACT

The novel anti-PD-L1/TGFBR2-ECD fusion protein (BR102) comprises an anti-PD-L1 antibody (HS636) which is fused at the C terminus of the heavy chain to a TGF-ß1 receptor Ⅱ ectodomain (TGFBR2-ECD), and which can sequester the PD-1/PD-L1 pathway and TGF-ß bioactivity in the immunosuppressive tumor microenvironment. In the expression of TGFBR2-ECD wild-type fused protein (BR102-WT), a 50 kDa clipped species was confirmed to be induced by proteolytic cleavage at a "QKS" site located in the N-terminus of the ectodomain, which resulted in the formation of IgG-like clipping. The matrix metalloproteinase-9 was determined to be associated with BR102-WT digestion. In addition, it was observed that the N-glycosylation modifications of the fusion protein were tightly involved in regulating proteolytic activity and the levels of cleavage could be significantly suppressed by MMP-inhibitors. To avoid proteolytic degradation, eliminating protease-sensitive amino acid motifs and introducing potential glycosylation were performed. Three sensitive motifs were mutated, and the levels of clipping were strongly restrained. The mutant candidates exhibited similar binding affinities to hPD-L1 and hTGF-ß1 as well as highly purified BR102-WT2. Furthermore, the mutants displayed more significant proteolytic resistance than that of BR102-WT2 in the lysate incubation reaction and the plasma stability test. Moreover, the bifunctional candidate Mu3 showed an additive antitumor effect in MC38/hPD-L1 bearing models as compared to that of with anti-PD-L1 antibody alone. In conclusion, in this study, the protease-sensitive features of BR102-WT were well characterized and efficient optimization was performed. The candidate BR102-Mutants exhibited advanced druggability in drug stability and displayed desirable antitumor activity.


Subject(s)
Antibodies, Neoplasm/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Colonic Neoplasms/therapy , Protein Processing, Post-Translational , Receptor, Transforming Growth Factor-beta Type II/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CHO Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cricetulus , Female , Glycosylation , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Mice , Mice, Inbred C57BL , Mutation , Protein Domains , Proteolysis , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
3.
J Clin Invest ; 131(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34720089

ABSTRACT

Evasion of the immune response is a hallmark of cancer, and programmed cell death 1 (PD-1) and PD-1 ligand 1 (PD-L1) are major mediators of this immunosuppression. Chitinase 3-like 1 (CHI3L1) is induced in many cancers, where it portends a poor prognosis and contributes to tumor metastasis and spread. However, the mechanism(s) that CHI3L1 uses in metastasis have not been defined. Here we demonstrate that CHI3L1 regulates the expression of PD-L1, PD-L2, PD-1, LAG3, and TIM3 and plays a critical role in melanoma progression and lymphatic spread. CHI3L1 also contributed to IFN-γ-stimulated macrophage PD-L1 expression, and RIG-like helicase innate immunity suppressed CHI3L1, PD-L1, and melanoma progression. Individual antibodies against CHI3L1 or PD-1 had discrete antitumor effects and additive antitumor responses in metastasis models and T cell-tumor cell cocultures when administered simultaneously. Synergistic cytotoxic tumor cell death was seen in T cell-tumor cell cocultures, and significantly enhanced antitumor responses were seen in in vivo tumor models treated with bispecific antibodies that simultaneously target CHI3L1 and PD-1. CHI3L1 contributes to tumor progression by stimulating the PD-1/PD-L1 axis and other checkpoint molecules. The simultaneous targeting of CHI3L1 and the PD-1/PD-L1 axis with individual and, more powerfully, with bispecific antibodies represents a promising therapy for pulmonary metastasis and progression.


Subject(s)
Antibodies, Bispecific , Antibodies, Neoplasm , B7-H1 Antigen , Chitinase-3-Like Protein 1 , Lung Neoplasms , Neoplasm Proteins , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Chitinase-3-Like Protein 1/antagonists & inhibitors , Chitinase-3-Like Protein 1/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology
4.
Br J Haematol ; 193(3): 581-591, 2021 05.
Article in English | MEDLINE | ID: mdl-33570193

ABSTRACT

Multiple functions of CD38 need exploring to expand clinical application of anti-CD38 antibodies in multiple myeloma (MM). We investigated membrane dynamics of MM cells and subsequent events when CD38 is targeted by therapeutic antibodies. Human MM cells (BF01) were co-cultured in vitro with therapeutic antibody (or control immunoglobulin G) and analysed using gene expression profiling. Microvesicles from antibody-exposed cells were analysed for differential gene and microRNA (miRNA) expression, and for phenotypic characterisation. Exposure of BF01 cells to anti-CD38 antibody resulted in CD38 membrane redistribution, upregulation of metabolism-related genes and downregulation of genes involved in cell cycle processes. Microvesicles derived from antibody-exposed cells showed increased CD73 and CD39 expression, presence of programmed death-ligand 1 and significant up-/down-modulation of miRNAs. Microvesicles accumulated around immunoglobulin Fc receptor-positive (FcR+ ) cells. Upon internalisation, natural killer cells displayed significantly increased expression of genes related to activation and immune response, and downregulation of genes involved in the cell cycle. Cells may use microvesicles to transmit signals distally as part of a survival strategy. Microvesicles are equipped on their surface with enzymatic machinery leading to production of tolerogenic adenosine. Further, they are internalised in FcR+ cells with significant functional modifications. These observations have relevance for improving anti-CD38 therapeutic antibodies through targeting this mechanism and its sequelae.


Subject(s)
ADP-ribosyl Cyclase 1/biosynthesis , Antibodies, Neoplasm/pharmacology , Cell Membrane/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Membrane Glycoproteins/biosynthesis , Multiple Myeloma/metabolism , Neoplasm Proteins/biosynthesis , Cell Line, Tumor , Humans , MicroRNAs/biosynthesis , Multiple Myeloma/drug therapy , RNA, Neoplasm/biosynthesis
5.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33627407

ABSTRACT

Anticytotoxic T lymphocyte-associated protein 4 (CTLA4) antibodies have shown potent antitumor activity, but systemic immune activation leads to severe immune-related adverse events, limiting clinical usage. We developed novel, conditionally active biologic (CAB) anti-CTLA4 antibodies that are active only in the acidic tumor microenvironment. In healthy tissue, this binding is reversibly inhibited by a novel mechanism using physiological chemicals as protein-associated chemical switches (PaCS). No enzymes or potentially immunogenic covalent modifications to the antibody are required for activation in the tumor. The novel anti-CTLA4 antibodies show similar efficacy in animal models compared to an analog of a marketed anti-CTLA4 biologic, but have markedly reduced toxicity in nonhuman primates (in combination with an anti-PD1 checkpoint inhibitor), indicating a widened therapeutic index (TI). The PaCS encompass mechanisms that are applicable to a wide array of antibody formats (e.g., ADC, bispecifics) and antigens. Examples shown here include antibodies to EpCAM, Her2, Nectin4, CD73, and CD3. Existing antibodies can be engineered readily to be made sensitive to PaCS, and the inhibitory activity can be optimized for each antigen's varying expression level and tissue distribution. PaCS can modulate diverse physiological molecular interactions and are applicable to various pathologic conditions, enabling differential CAB antibody activities in normal versus disease microenvironments.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Colonic Neoplasms/therapy , Immunotherapy/methods , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Neoplasm/chemistry , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Bicarbonates/chemistry , CD3 Complex/antagonists & inhibitors , CD3 Complex/genetics , CD3 Complex/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/immunology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression , Humans , Hydrogen Sulfide/chemistry , Hydrogen-Ion Concentration , Macaca fascicularis , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Protein Engineering/methods , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
6.
Mol Cancer Ther ; 20(1): 121-131, 2021 01.
Article in English | MEDLINE | ID: mdl-33277440

ABSTRACT

TIGIT is an immune checkpoint inhibitor expressed by effector CD4+ and CD8+ T cells, NK cells, and regulatory T cells (Tregs). Inhibition of TIGIT-ligand binding using antagonistic anti-TIGIT mAbs has shown in vitro potential to restore T-cell function and therapeutic efficacy in murine tumor models when combined with an anti-PD(L)-1 antibody. In the current work, we demonstrate broader TIGIT expression than previously reported in healthy donors and patients with cancer with expression on γδ T cells, particularly in CMV-seropositive donors, and on tumor cells from hematologic malignancies. Quantification of TIGIT density revealed tumor-infiltrating Tregs as the population expressing the highest receptor density. Consequently, the therapeutic potential of anti-TIGIT mAbs might be wider than the previously described anti-PD(L)-1-like restoration of αß T-cell function. CD155 also mediated inhibition of γδ T cells, an immune population not previously described to be sensitive to TIGIT inhibition, which could be fully prevented via use of an antagonistic anti-TIGIT mAb (EOS-448). In PBMCs from patients with cancer, as well as in tumor-infiltrating lymphocytes from mice, the higher TIGIT expression in Tregs correlated with strong antibody-dependent killing and preferential depletion of this highly immunosuppressive population. Accordingly, the ADCC/ADCP-enabling format of the anti-TIGIT mAb had superior antitumor activity, which was dependent upon Fcγ receptor engagement. In addition, the anti-TIGIT mAb was able to induce direct killing of TIGIT-expressing tumor cells both in human patient material and in animal models, providing strong rationale for therapeutic intervention in hematologic malignancies. These findings reveal multiple therapeutic opportunities for anti-TIGIT mAbs in cancer therapeutics.


Subject(s)
Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , Cytotoxicity, Immunologic , Receptors, Immunologic/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD/metabolism , Cytotoxicity, Immunologic/drug effects , Female , Healthy Volunteers , Humans , Immunoglobulin G/metabolism , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, IgG/metabolism , Receptors, Immunologic/metabolism , T-Lymphocytes, Regulatory/drug effects
7.
J Immunol ; 205(8): 2156-2168, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32887749

ABSTRACT

The T cell immunoreceptor with Ig and ITIM domains (TIGIT) has been shown to exert inhibitory roles in antitumor immune responses. In this study, we report the development of a human mAb, T4, which recognizes both human and mouse TIGIT and blocks the interaction of TIGIT with its ligand CD155 in both species. The T4 Ab targets the segment connecting F and G strands of TIGIT's extracellular IgV domain, and we show in studies with mouse tumor models that the T4 Ab exerts strong antitumor activity and induces durable immune memory against various tumor types. Mechanistically, we demonstrate that the T4 Ab's antitumor effects are mediated via multiple immunological impacts, including a CD8+ T immune response and Fc-mediated effector functions, through NK cells that cause significant reduction in the frequency of intratumoral T regulatory cells (Tregs). Notably, this Treg reduction apparently activates additional antitumor CD8+ T cell responses, targeting tumor-shared Ags that are normally cryptic or suppressed by Tregs, thus conferring cross-tumor immune memory. Subsequent engineering for Fc variants of the T4 Ab with enhanced Fc-mediated effector functions yielded yet further improvements in antitumor efficacy. Thus, beyond demonstrating the T4 Ab as a promising candidate for the development of cancer immunotherapies, our study illustrates how the therapeutic efficacy of an anti-TIGIT Ab can be improved by enhancing Fc-mediated immune effector functions. Our insights about the multiple mechanisms of action of the T4 Ab and its Fc variants should help in developing new strategies that can realize the full clinical potential of anti-TIGIT Ab therapies.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Neoplasm/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental , Receptors, Immunologic/antagonists & inhibitors , Animals , Antibodies, Blocking/immunology , Antibodies, Neoplasm/immunology , Antineoplastic Agents, Immunological/immunology , Female , Humans , Immunoglobulin Fc Fragments/immunology , Jurkat Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/immunology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Receptors, Immunologic/immunology , Xenograft Model Antitumor Assays
8.
Signal Transduct Target Ther ; 5(1): 141, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32855386

ABSTRACT

Programmed death ligand 1 (PD-L1) immune checkpoint inhibitors are promising therapeutic agents for treating cancers but the response rate is <20%. Some chemotherapeutic drugs could also activate an anticancer immune response to kill cancer cells, apart from their direct cytotoxicity. Our study investigated the combination of chemotherapeutic drugs with PD-L1 antibody to enhance the response rate of PD-L1 blockade. Non-small cell lung cancer (NSCLC) cells were pre-treated with mitomycin C (MMC) and then co-cultured with peripheral blood mononuclear cells (PBMCs) to investigate the effect of the combination of MMC with PD-L1 antibody. The drug combination was also evaluated in vivo in Lewis lung cancer (LLC) cells-bearing C57BL/6 mice. MMC increased the expressions of PD-L1 and MHC-I in NSCLC cells in vitro and in vivo and enhanced the cytotoxic effect of lymphocytes on NSCLC in vitro. In LLC-bearing mouse model, the combination of MMC and PD-L1 antibody was found to be more effective in retarding tumor growth and prolonging overall survival than either single treatment alone, which was associated with increased lymphocyte infiltration and granzyme B release. Mechanistically, MMC activated the ERK pathway, which subsequently enhanced the binding of c-JUN to the PD-L1 promoter and recruited its co-factor STAT3 to increase PD-L1 expression. The upregulated ERK pathway was shown to activate p65 to increase the MHC-I expression. MMC was shown to enhance the efficacy of PD-L1 blockade in NSCLC cells. Further study is warranted to translate the findings to clinical application.


Subject(s)
Antibodies, Neoplasm/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mitomycin/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , A549 Cells , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Human Umbilical Vein Endothelial Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Programmed Cell Death 1 Receptor/metabolism
9.
Anal Cell Pathol (Amst) ; 2020: 9582731, 2020.
Article in English | MEDLINE | ID: mdl-32802733

ABSTRACT

Nanosecond pulsed electric field (NsPEF) ablation effectively eliminates early-stage hepatocellular carcinoma (HCC) by local ablation and advanced HCC by inducing a remarkable and sustained host immune response. However, this approach is not sufficient to prevent cancer progression, and complementary approaches are necessary for effective immunotherapy. In this study, we evaluated the immunoactivating effects and mechanisms of action of nsPEF ablation and PD-1 blockade on an HCC orthotopic xenograft mouse model. Briefly, 24 C57BL-6J tumor-bearing mice were randomly assigned to three groups: nsPEF ablation group, anti-PD-1 administration group, and untreated control group. Tumor-infiltrating T, B, and NK cell levels and plasma concentrations of Th1 (IL-2, IFN-γ, and TNF-α), Th2 (IL-4, IL-5, IL-6, and IL-10), Th9 (IL-9), and Th17 (IL-17A, IL-17F, IL-21, and IL-22) cytokines were evaluated. Both nsPEF ablation and anti-PD-1 treatment induced immune cell infiltration in local tumors and modulated cytokine levels in the peripheral blood, with distinct changes in the two treatment groups. Based on these findings, both nsPEF ablation and PD-1 antibody administration can trigger a local and systemic immune response in a partially complementary manner, and nsPEF ablation should be considered along with PD-1 blockade for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Electricity , Liver Neoplasms/immunology , Nanoparticles/chemistry , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Carcinoma, Hepatocellular/blood , Cell Line, Tumor , Cytokines/blood , Humans , Immunity/drug effects , Killer Cells, Natural/drug effects , Liver Neoplasms/blood , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors
10.
JCI Insight ; 5(12)2020 06 18.
Article in English | MEDLINE | ID: mdl-32427583

ABSTRACT

The CD47/signal regulatory protein α (Cd47/SIRPα)interaction provides a macrophage immune checkpoint pathway that plays a critical role in cancer immune evasion across multiple cancers. Here, we report the engineering of a humanized anti-SIRPα monoclonal antibody (1H9) for antibody target cancer therapy. 1H9 has broad activity across a wide range of SIRPα variants. Binding of 1H9 to SIRPα blocks its interaction with CD47, thereby promoting macrophage-mediated phagocytosis of cancer cells. Preclinical studies in vitro and in vivo demonstrate that 1H9 synergizes with other therapeutic antibodies to promote phagocytosis of tumor cells and inhibit tumor growth in both syngeneic and xenograft tumor models, leading to survival benefit. Thus, 1H9 can potentially act as a universal agent to enhance therapeutic efficacy when used in combination with most tumor-targeting antibodies. We report a comparison of anti-SIRPα and anti-CD47 antibodies in CD47/SIRPα double-humanized mice and found that 1H9 exhibits a substantially reduced antigen sink effect due to the limited tissue distribution of SIRPα expression. Toxicokinetic studies in nonhuman primates show that 1H9 is well tolerated, with no treatment-related adverse effects noted. These data highlight the clinical potential of 1H9 as a pan-therapeutic with the desired properties when used in combination with tumor-targeting antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Macrophages/drug effects , Phagocytosis/drug effects , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/pharmacology , Antigens, Differentiation/metabolism , Cell Line, Tumor , Female , Immunotherapy/methods , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phagocytosis/immunology , Receptors, Immunologic/immunology
11.
Nihon Yakurigaku Zasshi ; 155(3): 150-154, 2020.
Article in Japanese | MEDLINE | ID: mdl-32378633

ABSTRACT

Many strategies have been tried to produce monoclonal antibodies (mAbs); however, there have been several problems about focusing on molecular targets and screening methods. For instance, the high tumor/normal ratio of antigen expression using DNA microarray has been thought to be important when we determine the molecular targets for antibody-drug. Although many antigens are expressed highly in tumors, those antigens have been removed from the candidates of antibody-drug targets because they were also expressed in normal tissues. We recently established a novel technology to produce a cancer-specific monoclonal antibody (CasMab). The post-translational difference such as glycans can be utilized to produce the CasMab, although the protein possesses the same amino acid sequence in both cancer and normal cells. We have already produced CasMabs against several glycoproteins such as podoplanin, which is expressed in both cancer and normal cells. Those CasMabs possess antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) in vitro and anti-tumor effect in xenograft models in vivo. In conclusion, the CasMab technology is the platform to develop cancer-specific mAbs, which could attack only cancer cells without side effects.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Antigens, Neoplasm/immunology , Glycoproteins/immunology , Animals , Antibody-Dependent Cell Cytotoxicity , Humans , Xenograft Model Antitumor Assays
12.
J Biol Chem ; 295(22): 7774-7788, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32317280

ABSTRACT

Glioblastoma multiforme (GBM) is a malignant brain tumor with a poor prognosis resulting from tumor resistance to anticancer therapy and a high recurrence rate. Compelling evidence suggests that this is driven by subpopulations of cancer stem cells (CSCs) with tumor-initiating potential. ABC subfamily B member 5 (ABCB5) has been identified as a molecular marker for distinct subsets of chemoresistant tumor-initiating cell populations in diverse human malignancies. In the current study, we examined the potential role of ABCB5 in growth and chemoresistance of GBM. We found that ABCB5 is expressed in primary GBM tumors, in which its expression was significantly correlated with the CSC marker protein CD133 and with overall poor survival. Moreover, ABCB5 was also expressed by CD133-positive CSCs in the established human U-87 MG, LN-18, and LN-229 GBM cell lines. Antibody- or shRNA-mediated functional ABCB5 blockade inhibited proliferation and survival of GBM cells and sensitized them to temozolomide (TMZ)-induced apoptosis in vitro Likewise, in in vivo human GBM xenograft experiments with immunodeficient mice, mAb treatment inhibited growth of mutant TP53, WT PTEN LN-229 tumors, and sensitized LN-229 tumors to TMZ therapy. Mechanistically, we demonstrate that ABCB5 blockade inhibits TMZ-induced G2/M arrest and augments TMZ-mediated cell death. Our results identify ABCB5 as a GBM chemoresistance marker and point to the potential utility of targeting ABCB5 to improve current GBM therapies.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Antibodies, Neoplasm/pharmacology , Apoptosis/drug effects , Brain Neoplasms , Drug Resistance, Neoplasm/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Glioblastoma , M Phase Cell Cycle Checkpoints/drug effects , Neoplasm Proteins , RNA, Small Interfering , Temozolomide/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
13.
Sci Adv ; 6(10): eaaz4204, 2020 03.
Article in English | MEDLINE | ID: mdl-32181368

ABSTRACT

Currently, there is a huge demand to develop chemoimmunotherapy with reduced systemic toxicity and potent efficacy to combat late-stage cancers with spreading metastases. Here, we report several "cocktail" therapeutic formulations by mixing immunogenic cell death (ICD)-inducing chemotherapeutics and immune adjuvants together with alginate (ALG) for localized chemoimmunotherapy. Immune checkpoint blockade (ICB) antibody may be either included into this cocktail for local injection or used via conventional intravenous injection. After injection of such cocktail into a solid tumor, in-situ gelation of ALG would lead to local retention and sustained release of therapeutics to reduce systemic toxicity. The chemotherapy-induced ICD with the help of immune adjuvant would trigger tumor-specific immune responses, which are further amplified by ICB to elicit potent systemic antitumor immune responses in destructing local tumors, eliminating metastases and inhibiting cancer recurrence. Our strategy of combining clinically used agents for tumor-localized cocktail chemoimmunotherapy possesses great potential for clinical translation.


Subject(s)
Antibodies, Neutralizing/pharmacology , Colonic Neoplasms/therapy , Combined Modality Therapy/methods , Doxorubicin/pharmacology , Mammary Neoplasms, Animal/therapy , Oxaliplatin/pharmacology , Adjuvants, Immunologic/administration & dosage , Alginates/chemistry , Animals , Antibodies, Neoplasm/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gels , Humans , Imiquimod/administration & dosage , Immunotherapy/methods , Injections, Intralesional , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
Blood ; 136(2): 210-223, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32219444

ABSTRACT

Resistance to multimodal chemotherapy continues to limit the prognosis of acute lymphoblastic leukemia (ALL). This occurs in part through a process called adhesion-mediated drug resistance, which depends on ALL cell adhesion to the stroma through adhesion molecules, including integrins. Integrin α6 has been implicated in minimal residual disease in ALL and in the migration of ALL cells to the central nervous system. However, it has not been evaluated in the context of chemotherapeutic resistance. Here, we show that the anti-human α6-blocking Ab P5G10 induces apoptosis in primary ALL cells in vitro and sensitizes primary ALL cells to chemotherapy or tyrosine kinase inhibition in vitro and in vivo. We further analyzed the underlying mechanism of α6-associated apoptosis using a conditional knockout model of α6 in murine BCR-ABL1+ B-cell ALL cells and showed that α6-deficient ALL cells underwent apoptosis. In vivo deletion of α6 in combination with tyrosine kinase inhibitor (TKI) treatment was more effective in eradicating ALL than treatment with a TKI (nilotinib) alone. Proteomic analysis revealed that α6 deletion in murine ALL was associated with changes in Src signaling, including the upregulation of phosphorylated Lyn (pTyr507) and Fyn (pTyr530). Thus, our data support α6 as a novel therapeutic target for ALL.


Subject(s)
Drug Resistance, Neoplasm , Gene Deletion , Integrin alpha6 , Neoplasm Proteins , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Pyrimidines/pharmacology , Animals , Antibodies, Neoplasm/pharmacology , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Integrin alpha6/genetics , Integrin alpha6/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy
15.
Asian Pac J Cancer Prev ; 21(2): 439-447, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32102522

ABSTRACT

OBJECTIVE: Human epidermal growth factor receptor 3 (HER3) is a unique member of the tyrosine kinase receptors with an inactive kinase domain and is the preferable dimerization partner for HER2 which lead to potent tumorigenic signaling. METHODS: In this study, the expression plasmids coding for the human HER3 subdomains were transfected into CHO-K1 cells. Produced proteins were characterized by ELISA and SDS-PAGE. Rabbits were immunized and produced polyclonal antibodies (pAbs) that were characterized by ELISA, Immunoblotting and flowcytometry and their inhibitory effects were assessed by XTT on BT-474 and JIMT-1 breast cancer cell lines. RESULT: The recombinant subdomains were highly immunogenic in rabbits. The pAbs reacted with the recombinant subdomains as well as commercial HER3 and the native receptor on tumor cell membranes and could significantly inhibit growth of Trastuzumab sensitive (BT-474) and resistant (JIMT-1) breast cancer cell lines in vitro. CONCLUSION: It seems that HER3 extra cellular domains (ECD) induce a strong anti-tumor antibody response and may prove to be potentially useful for immunotherapeutic applications.
.


Subject(s)
Antibodies, Neoplasm/immunology , Breast Neoplasms/immunology , Protein Domains/immunology , Receptor, ErbB-3/immunology , Animals , Antibodies, Neoplasm/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CHO Cells , Cell Line, Tumor , Cricetulus , Dimerization , Drug Resistance, Neoplasm , Humans , Immunization, Secondary , Immunoglobulin G , In Vitro Techniques , Mice , Plasmids , Rabbits , Receptor, ErbB-2/metabolism , Transfection , Vaccination
16.
Nanoscale ; 12(3): 1875-1885, 2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31903467

ABSTRACT

Tumor associated macrophages (TAMs) play an important role in initiating the immunosuppressive environment that negatively impacts the immunotherapy efficacy and has long been linked with cancer progression. On the other hand, activated macrophages display immense phagocytic potential and can be used as an effector cell for cancer therapy. But, activating TAMs to effectively phagocytose cancer cells is challenging. Cancer cells upregulate CD47, a "don't eat me" receptor that ligates with SIRPα present on macrophages to downregulate the phagocytosis. Since phagocytosis is a physical phenomenon based on engulfment of aberrant cells, we hypothesized that the phagocytic function of macrophages can be enhanced by blocking both CD47 and SIRPα in tandem and at the same time, engaging both macrophages and cancer cells can favor increased macrophage-cancer cellular interactions. Here, we demonstrate that a simple approach of anti-CD47 and anti-SIRPα antibodies conjugated lipid-based phagocytosis nanoenhancer (LPN) can perform both of these functions. The LPNs were stable in both physiological and biologically relevant conditions, bound to both macrophages and cancer cells and significantly enhanced phagocytosis of cancer cells as compared to combination of free antibodies. LPN treatment showed significant tumor growth inhibition and increased survival in B16F10 melanoma tumor bearing mice with no systemic toxicity. Mechanistic analysis of efficacy revealed an increase in intra-tumoral infiltration of effector T cells and NK cells. Cytokine analysis revealed increased secretion of intracellular iNOS, a hallmark of activated macrophages. This study shows that LPN can simultaneously block both CD47 and SIRPα and can effectively engage macrophage and cancer cell in close proximity. Combining these facets provide a simple approach to enhance phagocytosis and improve anti-cancer macrophage immunotherapy.


Subject(s)
Antibodies, Neoplasm , Immunotherapy , Lipids , Macrophages , Melanoma, Experimental , Nanoparticles , Phagocytosis , Animals , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/pharmacology , Humans , Lipids/chemistry , Lipids/pharmacology , Macrophages/immunology , Macrophages/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use
17.
JCI Insight ; 5(2)2020 01 30.
Article in English | MEDLINE | ID: mdl-31855578

ABSTRACT

Anti-programmed cell death protein 1 (anti-PD-1) therapy has become an immunotherapeutic backbone for treating many cancer types. Although many studies have aimed to characterize the immune response to anti-PD-1 therapy in the tumor and in the peripheral blood, relatively less is known about the changes in the tumor-draining lymph nodes (TDLNs). TDLNs are primary sites of tumor antigen exposure that are critical to both regulation and cross-priming of the antitumor immune response. We used multipanel mass cytometry to obtain a high-parameter proteomic (39 total unique markers) immune profile of the TDLNs in a well-studied PD-1-responsive, immunocompetent mouse model. Based on combined hierarchal gating and unsupervised clustering analyses, we found that anti-PD-1 therapy enhances remodeling of both B and T cell compartments toward memory phenotypes. Functionally, expression of checkpoint markers was increased in conjunction with production of IFN-γ, TNF-α, or IL-2 in key cell types, including B and T cell subtypes, and rarer subsets, such as Tregs and NKT cells. A deeper profiling of the immunologic changes that occur in the TDLN milieu during effective anti-PD-1 therapy may lead to the discovery of novel biomarkers for monitoring response and provide key insights toward developing combination immunotherapeutic strategies.


Subject(s)
Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , B-Lymphocytes/immunology , Lymph Nodes/drug effects , Neoplasms/immunology , Programmed Cell Death 1 Receptor/drug effects , T-Lymphocytes/immunology , Animals , Antibodies, Neoplasm/therapeutic use , Female , Interferon-gamma/metabolism , Interleukin-2/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Natural Killer T-Cells , Neoplasms/drug therapy , Proteomics , Tumor Necrosis Factor-alpha/metabolism
18.
J Immunol ; 203(7): 2011-2019, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31434709

ABSTRACT

OX40 is a costimulatory molecule from the TNFR family. In mice, it is expressed on Foxp3+ regulatory T cells (Tregs) constitutively and on conventional CD4 (Tconv) and CD8 T cells after Ag encounter. OX40 agonists are in clinical development to enhance antitumor immune responses, and one proposed mechanism of action is loss of Treg suppressive function. Studies have postulated that agonist OX40 therapy can impair Treg suppressive function. Using tools developed since the initial studies were published, we evaluated a direct effect of OX40 agonism on Treg function. We conclude that OX40 agonist Abs do not intrinsically impair Treg function but rather enhance Tconv cell IL-2 production, increasing Treg and Tconv cell proliferation. OX40-stimulated Tregs retain suppressive function, but also gain IFN-γ, TNF-α, and granzyme B expression. These data help resolve mechanistic questions regarding OX40 agonist immunotherapy and thus are relevant to developing combination therapies that target distinct T cell functions.


Subject(s)
Antibodies, Neoplasm/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Neoplasm Proteins , Neoplasms , Receptors, OX40 , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Neoplasm/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Mice , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Receptors, OX40/antagonists & inhibitors , Receptors, OX40/genetics , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/pathology
19.
J Clin Invest ; 129(9): 3952-3962, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31424423

ABSTRACT

Sialyl Lewis A (sLeA, also known as CA19-9), a tetrasaccharide selectively and highly expressed on advanced adenocarcinomas including colon, stomach, and pancreatic cancers, has long been considered as an attractive target for active and passive vaccination. While progress in antibodies targeting tumor-associated protein antigens resulted in an impressive array of therapeutics for cancer treatment, similar progress in exploiting tumor-associated carbohydrate antigens, such as sLeA, has been hampered by the lack of a detailed understanding of the singular characteristics of these antigens. We have addressed this issue by analyzing antibodies derived from patients immunized with an sLeA/KLH vaccine. These antibodies were engineered to mediate tumor clearance in vivo in preclinical models through Fc-FcγR interactions. However, in contrast to protein antigens in which hFcγRIIIA engagement was both necessary and sufficient to mediate tumor clearance in both preclinical and clinical settings, a similar selective dependence was not seen for anti-sLeA antibodies. Thus, re-engineering the Fc portion of sLeA-targeting antibodies to broadly enhance their affinity for activating FcγRs led to an enhanced therapeutic effect. These findings will facilitate the development of more efficient anticancer therapies and further advance this promising class of therapeutic antibodies into clinical use.


Subject(s)
Antibodies, Neoplasm , CA-19-9 Antigen/immunology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Receptors, IgG/immunology , Animals , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , Cell Line, Tumor , Humans , Mice , Mice, Transgenic , Neoplasms, Experimental/pathology
20.
Glycoconj J ; 36(5): 399-408, 2019 10.
Article in English | MEDLINE | ID: mdl-31267246

ABSTRACT

Even though a vaccine that targets tumor-associated carbohydrate antigens on epithelial carcinoma cells presents an attractive therapeutic approach, relatively poor immunogenicity limits its development. In this study, we investigated the immunological activity of a fluoro-substituted Sialyl-Tn (F-STn) analogue coupled to the non-toxic cross-reactive material of diphtheria toxin197 (CRM197). Our results indicate that F-STn-CRM197 promotes a greater immunogenicity than non-fluorinated STn-CRM197. In the presence or absence of adjuvant, F-STn-CRM197 remarkably enhances both cellular and humoral immunity against STn by increasing antigen-specific lymphocyte proliferation and inducing a mixed Th1/Th2 response leading to production of IFN-γ and IL-4 cytokines, as well as STn-specific antibodies. Furthermore, antisera produced from F-STn-CRM197 immunization significantly recognizes STn-positive tumor cells and increases cancer cell lysis induced by antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) pathways. Our data suggest that this F-STn vaccine may be useful for cancer immunotherapy and possibly for prophylactic prevention of cancer.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antigens, Tumor-Associated, Carbohydrate/chemistry , Bacterial Proteins/pharmacology , Cancer Vaccines/pharmacology , Colonic Neoplasms/therapy , Glycoconjugates/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neoplasm/isolation & purification , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, Tumor-Associated, Carbohydrate/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Cancer Vaccines/chemical synthesis , Cancer Vaccines/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Female , Gene Expression , Glycoconjugates/chemical synthesis , Glycoconjugates/immunology , Halogenation , Humans , Immune Sera/chemistry , Immune Sera/pharmacology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Immunization , Immunogenicity, Vaccine , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Spleen/drug effects , Spleen/immunology , Th1-Th2 Balance
SELECTION OF CITATIONS
SEARCH DETAIL
...