Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.593
Filter
2.
Am J Clin Pathol ; 157(4): 494-497, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34643211

ABSTRACT

OBJECTIVES: To compare flow cytometric minimal residual disease (MRD) data obtained using the EuroFlow approach, including the CD38-multiepitope (ME) antibody or the VS38c antibody. METHODS: We evaluated 29 bone marrow samples from patients with multiple myeloma (MM), of whom 15 had received daratumumab within the past 6 months. We evaluated MRD data and fluorescence intensities. RESULTS: Qualitative MRD data were 100% concordant between the 2 approaches. In MRD-positive samples (n = 14), MRD levels showed an excellent correlation (R2 = 0.999). Whereas VS38c staining was strong in both normal plasma cells and MM cells, independent of daratumumab treatment, staining intensities for CD38 were lower in MM cells compared with normal plasma cells, and on both cell types CD38 expression was significantly reduced in daratumumab-treated patients. CONCLUSIONS: Both CD38-ME and VS38c allow reliable MRD detection in MM patients, but the high expression of VS38c allows easier identification of MM cells, especially in daratumumab-treated patients.


Subject(s)
ADP-ribosyl Cyclase 1 , Antibodies, Neoplasm , Membrane Glycoproteins , Multiple Myeloma , ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/immunology , Cell Count , Epitopes , Flow Cytometry , Humans , Membrane Glycoproteins/immunology , Multiple Myeloma/diagnosis , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Neoplasm, Residual/diagnosis , Neoplasm, Residual/immunology , Plasma Cells
3.
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
5.
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
6.
Elife ; 102021 10 05.
Article in English | MEDLINE | ID: mdl-34608861

ABSTRACT

Myeloid, T, and NK cells are key players in the elimination phase of cancer immunoediting, also referred to as cancer immunosurveillance. However, the role of B cells and NAbs, which are present prior to the encounter with cognate antigens, has been overlooked. One reason is due to the popular use of a single B cell-deficient mouse model, muMT mice. Cancer models using muMT mice display a similar tumor burden as their wild-type (WT) counterparts. Empirically, we observe what others have previously reported with muMT mice. However, using two other B cell-deficient mouse models (IgHELMD4 and CD19creDTA), we show a three- to fivefold increase in tumor burden relative to WT mice. In addition, using an unconventional, non-cancer-related, immune neoantigen model where hypoxic conditions and cell clustering are absent, we provide evidence that B cells and their innate, natural antibodies (NAbs) are critical for the detection and elimination of neoantigen-expressing cells. Finally, we find that muMT mice display anti-tumor immunity because of an unexpected compensatory mechanism consisting of significantly enhanced type 1 interferon (IFN)-producing plasmacytoid dendritic cells (pDCs), which recruit a substantial number of NK cells to the tumor microenvironment compared to WT mice. Diminishing this compensatory pDC-IFN-NK cell mechanism revealed that muMT mice develop a three- to fivefold increase in tumor burden compared to WT mice. In summary, our findings suggest that NAbs are part of an early defense against not only microorganisms and dying cells, but precancerous cells as well.


Subject(s)
Antibodies, Neoplasm/immunology , B-Lymphocytes/immunology , Immunity, Innate/genetics , Animals , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Interferon Type I/metabolism , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Microenvironment
7.
Front Immunol ; 12: 703574, 2021.
Article in English | MEDLINE | ID: mdl-34539634

ABSTRACT

CD38 is the major NAD+-hydrolyzing ecto-enzyme in most mammals. As a type II transmembrane protein, CD38 is also a promising target for the immunotherapy of multiple myeloma (MM). Nanobodies are single immunoglobulin variable domains from heavy chain antibodies that naturally occur in camelids. Using phage display technology, we isolated 13 mouse CD38-specific nanobodies from immunized llamas and produced these as recombinant chimeric mouse IgG2a heavy chain antibodies (hcAbs). Sequence analysis assigned these hcAbs to five distinct families that bind to three non-overlapping epitopes of CD38. Members of families 4 and 5 inhibit the GDPR-cyclase activity of CD38. Members of families 2, 4 and 5 effectively induce complement-dependent cytotoxicity against CD38-expressing tumor cell lines, while all families effectively induce antibody dependent cellular cytotoxicity. Our hcAbs present unique tools to assess cytotoxicity mechanisms of CD38-specific hcAbs in vivo against tumor cells and potential off-target effects on normal cells expressing CD38 in syngeneic mouse tumor models, i.e. in a fully immunocompetent background.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neoplasm/immunology , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Heavy Chains/immunology , Membrane Glycoproteins/immunology , Neoplasms/immunology , ADP-ribosyl Cyclase 1/genetics , Animals , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Humans , Immunoglobulin Heavy Chains/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout
8.
Mol Med Rep ; 24(5)2021 Nov.
Article in English | MEDLINE | ID: mdl-34490477

ABSTRACT

Phage display technology (PD) is a powerful technique for the generation of tumor­targeting antibodies. However, there are a number of different selection methods established in different laboratories around the world. Cell­based PD panning methods using primary tumor cells are particularly heterogeneous between laboratories, which can lead to inconsistent results. Therefore, the present study evaluated different cell­based PD selection methods regarding their potential to generate acute myeloid leukemia (AML) blast­binding antibodies. In addition to this evaluation, the present study improved the PD procedure by optimizing selection as well as depletion strategies. To the best of our knowledge, the current study demonstrated for the first time that antigen diversity during the depletion step is of importance for the enrichment of tumor­targeting phage antibodies. It is demonstrated that medium levels of depletion antigen diversity led to the most promising antibody candidates. In addition, it was determined that purification of blast cells from patients with AML by immunomagnetic separation ameliorated the selection of AML­binding phages during panning. Furthermore, suggesting a common design­related mechanism using a 'single­pot' PD library, such as the well­known Tomlinson single­chain fragment variable (scFv) library, the present study identified specific binding consensus phage particles in independent panning procedures. By means of these optimized strategies, four promising AML blast­binding phage particles were isolated and soluble scFv­Fc (scFv cloned to a fragment crystallizable of an IgG2a mouse antibody) fusion proteins were produced. These scFv­Fc antibodies bound the surface of AML blasts and were successfully internalized into their cytoplasm, indicating that they are potential immunoconjugate candidates for AML immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Cell Surface Display Techniques/methods , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Antibody Specificity , Bacteriophages/immunology , HEK293 Cells , Humans , Primary Cell Culture
9.
Plast Reconstr Surg ; 148(2): 287-298, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34398081

ABSTRACT

BACKGROUND: Women with cosmetic breast implants have significantly lower rates of subsequent breast cancer than the general population (relative risk, 0.63; 95 percent CI, 0.56 to 0.71). The authors hypothesize that breast implant-induced local inflammation stimulates immunosurveillance recognition of breast tumor antigen. METHODS: Sera were collected from two cohorts of healthy women: women with long-term breast implants (i.e., breast implants for >6 months) and breast implant-naive women. Antibody responses to breast tumor antigens were tested by enzyme-linked immunosorbent assay and compared between cohorts by unpaired t test. Of the implant-naive cohort, nine women underwent breast augmentation, and antibody responses before and after implant placement were compared by paired t test. RESULTS: Sera were collected from 104 women: 36 (34.6 percent) long-term breast implants and 68 (65.4 percent) implant-naive women. Women with long-term breast implants had higher antibody responses than implant-naive women to mammaglobin-A (optical density at 450 nm, 0.33 versus 0.22; p = 0.003) and mucin-1 (optical density at 450 nm, 0.42 versus 0.34; p = 0.02). There was no difference in antibody responses to breast cancer susceptibility gene 2, carcinoembryonic antigen, human epidermal growth factor receptor-2, or tetanus. Nine women with longitudinal samples preoperatively and 1 month postoperatively demonstrated significantly elevated antibody responses following implant placement to mammaglobin-A (mean difference, 0.13; p = 0.0002) and mucin-1 (mean difference 0.08; p = 0.02). There was no difference in postimplant responses to other breast tumor antigens, or tetanus. CONCLUSIONS: Women with long-term breast implants have higher antibody recognition of mammaglobin-A and mucin-1. This study provides the first evidence of implant-related immune responses to breast cancer antigens. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Subject(s)
Antibodies, Neoplasm/blood , Breast Implantation/instrumentation , Breast Implants , Breast Neoplasms/prevention & control , Immunologic Surveillance , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Case-Control Studies , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Healthy Volunteers , Humans , Middle Aged , Serologic Tests/statistics & numerical data , Silicone Gels , Young Adult
10.
Cell Rep ; 36(1): 109309, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233193

ABSTRACT

αvß8 integrin, a key activator of transforming growth factor ß (TGF-ß), inhibits anti-tumor immunity. We show that a potent blocking monoclonal antibody against αvß8 (ADWA-11) causes growth suppression or complete regression in syngeneic models of squamous cell carcinoma, mammary cancer, colon cancer, and prostate cancer, especially when combined with other immunomodulators or radiotherapy. αvß8 is expressed at the highest levels in CD4+CD25+ T cells in tumors, and specific deletion of ß8 from T cells is as effective as ADWA-11 in suppressing tumor growth. ADWA-11 increases expression of a suite of genes in tumor-infiltrating CD8+ T cells normally inhibited by TGF-ß and involved in tumor cell killing, including granzyme B and interferon-γ. The in vitro cytotoxic effect of tumor CD8 T cells is inhibited by CD4+CD25+ cells, and this suppressive effect is blocked by ADWA-11. These findings solidify αvß8 integrin as a promising target for cancer immunotherapy.


Subject(s)
Immunity , Immunotherapy , Integrins/metabolism , Models, Biological , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Granzymes/metabolism , Interferon-gamma/metabolism , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction , Smad3 Protein/metabolism , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
11.
MAbs ; 13(1): 1951427, 2021.
Article in English | MEDLINE | ID: mdl-34291723

ABSTRACT

Antibody-drug conjugates (ADCs) are a rapidly expanding class of biotherapeutics that utilize antibodies to selectively deliver cytotoxic drugs to the tumor site. As of May 2021, the U.S. Food and Drug Administration (FDA) has approved ten ADCs, namely Adcetris®, Kadcyla®, Besponsa®, Mylotarg®, Polivy®, Padcev®, Enhertu®, Trodelvy®, Blenrep®, and Zynlonta™ as monotherapy or combinational therapy for breast cancer, urothelial cancer, myeloma, acute leukemia, and lymphoma. In addition, over 80 investigational ADCs are currently being evaluated in approximately 150 active clinical trials. Despite the growing interest in ADCs, challenges remain to expand their therapeutic index (with greater efficacy and less toxicity). Recent advances in the manufacturing technology for the antibody, payload, and linker combined with new bioconjugation platforms and state-of-the-art analytical techniques are helping to shape the future development of ADCs. This review highlights the current status of marketed ADCs and those under clinical investigation with a focus on translational strategies to improve product quality, safety, and efficacy.


Subject(s)
Antibodies, Neoplasm , Antineoplastic Agents, Immunological , Drug Delivery Systems , Immunoconjugates , Neoplasms , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/therapeutic use , Clinical Trials as Topic , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology
13.
Cell Rep ; 35(6): 109110, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979623

ABSTRACT

Antibody repertoire sequencing enables researchers to acquire millions of B cell receptors and investigate these molecules at the single-nucleotide level. This power and resolution in studying humoral responses have led to its wide applications. However, most of these studies were conducted with a limited number of samples. Given the extraordinary diversity, assessment of these key features with a large sample set is demanded. Thus, we collect and systematically analyze 2,152 high-quality heavy-chain antibody repertoires. Our study reveals that 52 core variable genes universally contribute to more than 99% of each individual's repertoire; a distal interspersed preferences characterize V gene recombination; the number of public clones between two repertoires follows a linear model, and the positive selection dominates at RGYW motif in somatic hypermutations. Thus, this population-level analysis resolves some critical features of the antibody repertoire and may have significant value to the large cadre of scientists.


Subject(s)
Antibodies, Neoplasm/immunology , Biology/methods , High-Throughput Nucleotide Sequencing/methods , Receptors, Antigen, B-Cell/metabolism , V(D)J Recombination/immunology , Datasets as Topic , Humans
14.
PLoS One ; 16(4): e0248903, 2021.
Article in English | MEDLINE | ID: mdl-33857179

ABSTRACT

Following curative immunotherapy of B16F10 tumors, ~60% of mice develop a strong antibody response against cell-surface tumor antigens. Their antisera confer prophylactic protection against intravenous challenge with B16F10 cells, and also cross-react with syngeneic and allogeneic tumor cell lines MC38, EL.4, 4T1, and CT26. We identified the envelope glycoprotein (env) of a murine endogenous retrovirus (ERV) as the antigen accounting for the majority of this humoral response. A systemically administered anti-env monoclonal antibody cloned from such a response protects against tumor challenge, and prophylactic vaccination against the env protein protects a majority of naive mice from tumor establishment following subcutaneous inoculation with B16F10 cells. These results suggest the potential for effective prophylactic vaccination against analogous HERV-K env expressed in numerous human cancers.


Subject(s)
Antibodies, Neoplasm/immunology , Endogenous Retroviruses/immunology , Gene Products, env/immunology , Immunotherapy/methods , Neoplasms , Animals , Cell Line, Tumor , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy
15.
Science ; 371(6533)2021 03 05.
Article in English | MEDLINE | ID: mdl-33649166

ABSTRACT

TP53 (tumor protein p53) is the most commonly mutated cancer driver gene, but drugs that target mutant tumor suppressor genes, such as TP53, are not yet available. Here, we describe the identification of an antibody highly specific to the most common TP53 mutation (R175H, in which arginine at position 175 is replaced with histidine) in complex with a common human leukocyte antigen-A (HLA-A) allele on the cell surface. We describe the structural basis of this specificity and its conversion into an immunotherapeutic agent: a bispecific single-chain diabody. Despite the extremely low p53 peptide-HLA complex density on the cancer cell surface, the bispecific antibody effectively activated T cells to lyse cancer cells that presented the neoantigen in vitro and in mice. This approach could in theory be used to target cancers containing mutations that are difficult to target in conventional ways.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , HLA-A2 Antigen/immunology , Neoplasms/therapy , Tumor Suppressor Protein p53/immunology , Alleles , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/therapeutic use , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/therapeutic use , Arginine/genetics , COS Cells , Chlorocebus aethiops , Female , HEK293 Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/genetics , Histidine/genetics , Humans , Immunization, Passive , Jurkat Cells , Lymphocyte Activation , Mice, Inbred NOD , Mutation , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
16.
Int J Nanomedicine ; 16: 403-420, 2021.
Article in English | MEDLINE | ID: mdl-33469292

ABSTRACT

BACKGROUND: Therapeutic tumor vaccines are one of the most promising strategies and have attracted great attention in cancer treatment. However, most of them have shown unsatisfactory immunogenicity, there are still few available vaccines for clinical use. Therefore, there is an urgent demand to develop novel strategies to improve the immune efficacy of antitumor vaccines. PURPOSE: This study aimed to develop novel adjuvants and carriers to enhance the immune effect of MUC1 glycopeptide antigen-based antitumor vaccines. METHODS: An antitumor vaccine was developed, in which MUC1 glycopeptide was used as tumor-associated antigen, α-GalCer served as an immune adjuvant and AuNPs was a multivalent carrier. RESULTS: Immunological evaluation results indicated that the constructed vaccines enabled a significant antibody response. FACS analysis and immunofluorescence assay showed that the induced antisera exhibited a specific binding with MUC1 positive MCF-7 cells. Moreover, the induced antibody can mediate CDC to kill MCF-7 cells. Besides stimulating B cells to produce MUC1-specific antibodies, the prepared vaccines also induced MUC1-specific CTLs in vitro. Furthermore, the vaccines significantly delayed tumor development in tumor-bearing mice model. CONCLUSION: These results showed that the construction of vaccines by presenting α-GalCer adjuvant and an antigen on gold nanoparticles offers a potential strategy to improve the antitumor response in cancer immunotherapy.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Galactosylceramides/pharmacology , Gold/pharmacology , Metal Nanoparticles/chemistry , Mucin-1/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neoplasm/immunology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Galactosylceramides/chemical synthesis , Galactosylceramides/chemistry , Humans , Immune Sera/metabolism , Melanoma/immunology , Melanoma/pathology , Metal Nanoparticles/ultrastructure , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
17.
Mol Cancer ; 20(1): 11, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33413365

ABSTRACT

Emerging evidence has revealed that the removal of N-linked glycosylation could enhance PD-L1 detection. However, whether PD-L1 antibodies against different epitopes of PD-L1 antigens responding to deglycosylation has not been characterized. In this study, we compared natural and deglycosylated PD-L1 expression in lung cancer (LuCa) using a panel of PD-L1 antibodies (28-8, CAL10, 73-10 and SP142). We found that removal of N-linked glycosylation markedly enhanced PD-L1 detection when the 28-8, CAL10 and SP142 monoclonal antibodies (mAbs) were used but slightly inhibited PD-L1 detection when the 73-10 mAb was used. Moreover, for the CAL10 and SP142 mAbs, deglycosylated PD-L1 levels showed stronger correlations with the response to anti-PD-1 therapy. Overall, our research provides a comprehensive insight into the application of deglycosylated PD-L1 detection, which expands the clinical significance of this established strategy in LuCa.


Subject(s)
B7-H1 Antigen/metabolism , Lung Neoplasms/metabolism , Antibodies, Neoplasm/immunology , B7-H1 Antigen/immunology , Glycosylation , Humans , Immunohistochemistry , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Tomography, X-Ray Computed , Treatment Outcome
18.
Front Immunol ; 12: 763877, 2021.
Article in English | MEDLINE | ID: mdl-35069536

ABSTRACT

Malignant melanoma is an aggressive form of cancer, which can be treated with anti-CTLA-4 and anti-PD-1 checkpoint inhibitor antibodies but while anti-CTLA-4 antibodies have clear benefits for some patients with melanoma, productive responses are difficult to predict and often associated with serious immune related adverse events. Antibodies specific to CTLA-4 bind two major isoforms of CTLA-4 in humans, the receptor isoform and a second naturally secretable, soluble isoform - sCTLA-4. The primary aim here was to examine the effect of selectively blocking the function of sCTLA-4 on in vitro immune responses from volunteer healthy or melanoma patient PBMC samples. Addition of recombinant sCTLA-4 to healthy PBMC samples demonstrated sCTLA-4 to have immunosuppressive capacity comparable to recombinant CTLA4-Ig, partially reversible upon antibody blockade. Further, we identified a mechanistic relationship where melanoma patient TGFß2 serum levels correlated with sCTLA-4 levels and provided the basis for a novel protocol to enhance sCTLA-4 production and secretion by T cells with TGFß2. Finally, a comparison of selective antibody blockade of sCTLA-4 demonstrated that both healthy and melanoma patient effector cytokine responses can be significantly increased. Overall, the data support the notion that sCTLA-4 is a contributory factor in cancer immune evasion.


Subject(s)
Antibodies, Neoplasm/immunology , CTLA-4 Antigen/immunology , Immune Checkpoint Inhibitors , Melanoma , Neoplasm Proteins/immunology , Transforming Growth Factor beta2/immunology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Humans , Male , Melanoma/immunology , Melanoma/therapy , Mice , Middle Aged
19.
Immunol Med ; 44(2): 69-73, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32693699

ABSTRACT

Chimeric antigen receptor (CAR) is generated by fusing a cancer-specific antibody's antigen recognition site with costimulatory molecules such as CD28 and CD3ζ. T cells transduced with CAR recognizes cancer-specific antigens and kill cancer cells. The effect of CD19-targeted CAR T cells on B-cell hematologic cancer is surprising and has already been approved in many countries including Japan. More targets for several kinds of cancers are being searched now. We have also shown that CAR T cells specific for activated integrin ß7 are highly effective for multiple myeloma in pre-clinical tests.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/therapeutic use , Antibodies, Neoplasm/immunology , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Humans , Integrin beta Chains , Multiple Myeloma/immunology , Multiple Myeloma/therapy , T-Lymphocytes/immunology
20.
Jpn J Clin Oncol ; 51(3): 408-415, 2021 Mar 03.
Article in English | MEDLINE | ID: mdl-33244584

ABSTRACT

OBJECTIVE: Fludarabine, cyclophosphamide and rituximab (FCR) is the standard regimen for fit patients with untreated CD20-positive chronic lymphocytic leukemia (CLL). However, this combination is unavailable in Japan because rituximab is not approved for CLL. We investigated the efficacy and safety of FCR in this single-arm, multicenter study designed as a bridging study to the CLL8 study by the German CLL Study Group. METHODS: The study enrolled previously untreated patients with CLL of Binet stage B or C with active disease. Patients with a Cumulative Illness Rating Scale score of ≤6 and creatinine clearance of ≥70 ml/min were eligible. Patients received 6 cycles of FCR every 28 days and were followed for up to 1 year. RESULTS: Seven patients were enrolled. The best overall response rate according to the 1996 NCI-WG Guidelines, the primary endpoint of the study, was 71.4% (95% confidence interval, 29.0-96.3%), with one patient achieving complete response. No deaths or progression occurred during follow-up. The main adverse event was hematotoxicity. CD4-positive T-cell count decreased in all patients; most patients showed no reduction in serum immunoglobulin G. CONCLUSION: Although the number of patients was limited, FCR appears to be effective with manageable toxicity for treatment-naïve fit Japanese patients with CD20-positive CLL. CLINICAL TRIAL NUMBER: JapicCTI-132285.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Rituximab/therapeutic use , Vidarabine/analogs & derivatives , Aged , Antibodies, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Cyclophosphamide/adverse effects , Cyclophosphamide/pharmacokinetics , Disease Progression , Endpoint Determination , Female , Humans , Immunosuppression Therapy , Japan , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Male , Middle Aged , Remission Induction , Rituximab/adverse effects , Rituximab/blood , Rituximab/pharmacokinetics , Treatment Outcome , Vidarabine/adverse effects , Vidarabine/pharmacokinetics , Vidarabine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...