Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.061
Filter
1.
Sci Rep ; 14(1): 9457, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658627

ABSTRACT

Increased use of therapeutic monoclonal antibodies and the relatively high manufacturing costs fuel the need for more efficient production methods. Here we introduce a novel, fast, robust, and safe isolation platform for screening and isolating antibody-producing cell lines using a nanowell chip and an innovative single-cell isolation method. An anti-Her2 antibody producing CHO cell pool was used as a model. The platform; (1) Assures the single-cell origin of the production clone, (2) Detects the antibody production of individual cells and (3) Isolates and expands the individual cells based on their antibody production. Using the nanowell platform we demonstrated an 1.8-4.5 increase in anti-Her2 production by CHO cells that were screened and isolated with the nanowell platform compared to CHO cells that were not screened. This increase was also shown in Fed-Batch cultures where selected high production clones showed titers of 19-100 mg/L on harvest day, while the low producer cells did not show any detectable anti-Her2 IgG production. The screening of thousands of single cells is performed under sterile conditions and the individual cells were cultured in buffers and reagents without animal components. The time required from seeding a single cell and measuring the antibody production to fully expanded clones with increased Her-2 production was 4-6 weeks.


Subject(s)
Antibodies, Monoclonal , Cricetulus , Receptor, ErbB-2 , CHO Cells , Animals , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/biosynthesis , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , Humans , Cell Separation/methods , Single-Cell Analysis/methods
2.
Eur J Drug Metab Pharmacokinet ; 49(3): 383-392, 2024 May.
Article in English | MEDLINE | ID: mdl-38564097

ABSTRACT

BACKGROUND AND OBJECTIVE: GB221 is a recombinant humanized anti-HER2 monoclonal antibody. The purpose of this study was to evaluate the pharmacokinetic, safety, and immunogenicity of GB221 in healthy Chinese adults in comparison to trastuzumab (Herceptin®). METHODS: In this randomized, double-blind, parallel-group phase I clinical trial, 88 subjects were randomized 1:1 to receive a single intravenous infusion (90-100 min) of GB221 or trastuzumab (6 mg/kg). The primary pharmacokinetic parameters-maximum observed serum concentration (Cmax), area under the serum concentration-time curve from zero to the last quantifiable concentration at time t (AUC0-t), and area under the serum concentration-time curve from time zero to infinity (AUC0-∞)-of GB221 and trastuzumab were compared to establish whether the 90% confidence interval (CI) attained the 80-125% bioequivalence standard. Safety and immunogenicity were also evaluated. RESULTS: The GB221 group (n = 43) and the trastuzumab group (n = 44) showed similar pharmacokinetic characteristics. The geometric mean ratios (90% CI) of Cmax, AUC0-t, and AUC0-∞ between the two groups were 107.53% (102.25-113.07%), 108.31% (103.57-113.26%), and 108.34% (103.57-113.33%), respectively. The incidence of treatment-emergent adverse events (TEAEs) was 83.7% (36/43) of the subjects in the GB221 group and 95.5% (42/44) of the subjects in the trastuzumab group. No subjects withdrew from the trial due to TEAEs, and there were no occurrences of serious adverse events. All subjects tested negative for antidrug antibodies (ADA). CONCLUSION: GB221 demonstrated similar pharmacokinetics to trastuzumab and comparable safety and immunogenicity in healthy Chinese adults.


Subject(s)
Antineoplastic Agents, Immunological , Area Under Curve , Therapeutic Equivalency , Trastuzumab , Humans , Trastuzumab/pharmacokinetics , Trastuzumab/administration & dosage , Trastuzumab/adverse effects , Adult , Male , Double-Blind Method , Female , Young Adult , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Asian People , Infusions, Intravenous , Middle Aged , Healthy Volunteers , Receptor, ErbB-2/immunology , East Asian People
3.
Breast Cancer Res ; 26(1): 40, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459598

ABSTRACT

BACKGROUND: 99mTc radiolabeled nanobody NM-02 (99mTc-NM-02) is a novel single photon emission computed tomography (SPECT) probe with a high affinity and specificity for human epidermal growth factor receptor 2 (HER2). In this study, a clinical imaging trial was conducted to investigate the relationship between 99mTc-NM-02 uptake and HER2 expression in patients with breast cancer. METHODS: Thirty patients with pathologically confirmed breast cancer were recruited and imaged with both 99mTc-NM-02 SPECT/computed tomography (CT) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/CT. According to the treatment conditions before recruitment, patients were divided into two groups, the newly diagnosed group (n = 24) and the treated group (n = 6). The maximal standard uptake value (SUVmax) of 18F-FDG and SUVmax and mean SUV (SUVmean) of 99mTc-NM-02 in the lesions were determined to analyze the relationship with HER2 expression. RESULTS: No meaningful relationship was observed between 18F-FDG uptake and HER2 expression in 30 patients with breast cancer. 99mTc-NM-02 uptake was positively correlated with HER2 expression in the newly diagnosed group, but no correlation was observed in the treated group. 99mTc-NM-02 uptake in HER2-positive lesions was lower in those with effective HER2-targeted therapy compared with the newly diagnosed group. 99mTc-NM-02 SPECT/CT detected brain and bone metastases of breast cancer with a different imaging pattern from 18F-FDG PET/CT. 99mTc-NM-02 showed no non-specific uptake in inflamed tissues and revealed intra- and intertumoral HER2 heterogeneity by SPECT/CT imaging in 9 of the 30 patients with breast cancer. CONCLUSIONS: 99mTc-NM-02 SPECT/CT has the potential for visualizing whole-body HER2 overexpression in untreated patients, making it a promising method for HER2 assessment in patients with breast cancer. TRIAL REGISTRATION: NCT04674722, Date of registration: December 19, 2020.


Subject(s)
Bone Neoplasms , Breast Neoplasms , Receptor, ErbB-2 , Female , Humans , Bone Neoplasms/secondary , Breast Neoplasms/diagnostic imaging , Fluorodeoxyglucose F18 , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/methods , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Single-Domain Antibodies
4.
Int J Mol Sci ; 25(3)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38339219

ABSTRACT

Monoclonal antibody (mAb)-based and/or cell-based immunotherapies provide innovative approaches to cancer treatments. However, safety concerns over targeting normal cells expressing reactive antigens still exist. Therefore, the development of cancer-specific mAbs (CasMabs) that recognize cancer-specific antigens with in vivo antitumor efficacy is required to minimize the adverse effects. We previously screened anti-human epidermal growth factor receptor 2 (HER2) mAbs and successfully established a cancer-specific anti-HER2 mAb, H2Mab-250/H2CasMab-2 (IgG1, kappa). In this study, we showed that H2Mab-250 reacted with HER2-positive breast cancer cells but did not show reactivity to normal epithelial cells in flow cytometry. In contrast, a clinically approved anti-HER2 mAb, trastuzumab, recognized both breast cancer and normal epithelial cells. We further compared the affinity, effector activation, and antitumor effect of H2Mab-250 with trastuzumab. The results showed that H2Mab-250 exerted a comparable antitumor effect with trastuzumab in the mouse xenograft models of BT-474 and SK-BR-3, although H2Mab-250 possessed a lower affinity and effector activation than trastuzumab in vitro. H2Mab-250 could contribute to the development of chimeric antigen receptor-T or antibody-drug conjugates without adverse effects for breast cancer therapy.


Subject(s)
Antibodies, Monoclonal , Antineoplastic Agents , Breast Neoplasms , Animals , Female , Humans , Mice , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , Heterografts , Receptor, ErbB-2/immunology , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 29(17): 3256-3258, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37428103

ABSTRACT

A vaccine targeting HER2, a nonmutated but overexpressed tumor antigen, readily primed T cells for ex vivo expansion and adoptive transfer with minimal toxicity. This regimen led to intramolecular epitope spreading in a majority of patients and offers a treatment modality that may improve outcomes for patients with metastatic breast cancer expressing HER2. See related article by Disis et al., p. 3362.


Subject(s)
Breast Neoplasms , Cancer Vaccines , Humans , Female , T-Lymphocytes/immunology , Breast Neoplasms/pathology , Cancer Vaccines/therapeutic use , Cancer Vaccines/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Antigens, Neoplasm/immunology
6.
JAMA Oncol ; 9(4): 490-499, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36602784

ABSTRACT

Importance: Both tumor-infiltrating lymphocytes (TILs) assessment and immune-related gene expression signatures by RNA profiling predict higher pathologic complete response (pCR) and improved event-free survival (EFS) in patients with early-stage ERBB2/HER2-positive breast cancer. However, whether these 2 measures of immune activation provide similar or additive prognostic value is not known. Objective: To examine the prognostic ability of TILs and immune-related gene expression signatures, alone and in combination, to predict pCR and EFS in patients with early-stage ERBB2/HER2-positive breast cancer treated in 2 clinical trials. Design, Setting, and Participants: In this prognostic study, a correlative analysis was performed on the Cancer and Leukemia Group B (CALGB) 40601 trial and the PAMELA trial. In the CALGB 40601 trial, 305 patients were randomly assigned to weekly paclitaxel with trastuzumab, lapatinib, or both for 16 weeks. The primary end point was pCR, with a secondary end point of EFS. In the PAMELA trial, 151 patients received neoadjuvant treatment with trastuzumab and lapatinib for 18 weeks. The primary end point was the ability of the HER2-enriched subtype to predict pCR. The studies were conducted from October 2013 to November 2015 (PAMELA) and from December 2008 to February 2012 (CALGB 40601). Data analyses were performed from June 1, 2020, to January 1, 2022. Main Outcomes and Measures: Immune-related gene expression profiling by RNA sequencing and TILs were assessed on 230 CALGB 40601 trial pretreatment tumors and 138 PAMELA trial pretreatment tumors. The association of these biomarkers with pCR (CALGB 40601 and PAMELA) and EFS (CALGB 40601) was studied by logistic regression and Cox analyses. Results: The median age of the patients was 50 years (IQR, 42-50 years), and 305 (100%) were women. Of 202 immune signatures tested, 166 (82.2%) were significantly correlated with TILs. In both trials combined, TILs were significantly associated with pCR (odds ratio, 1.01; 95% CI, 1.01-1.02; P = .02). In addition to TILs, 36 immune signatures were significantly associated with higher pCR rates. Seven of these signatures outperformed TILs for predicting pCR, 6 of which were B-cell related. In a multivariable Cox model adjusted for clinicopathologic factors, including PAM50 intrinsic tumor subtype, the immunoglobulin G signature, but not TILs, was independently associated with EFS (immunoglobulin G signature-adjusted hazard ratio, 0.63; 95% CI, 0.42-0.93; P = .02; TIL-adjusted hazard ratio, 1.00; 95% CI, 0.98-1.02; P = .99). Conclusions and Relevance: Results of this study suggest that multiple B-cell-related signatures were more strongly associated with pCR and EFS than TILs, which largely represent T cells. When both TILs and gene expression are available, the prognostic value of immune-related signatures appears to be superior.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , Lymphocytes, Tumor-Infiltrating , Receptor, ErbB-2 , Adult , Female , Humans , Middle Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Biomarkers, Tumor/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Immunoglobulin G/immunology , Lapatinib/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Neoadjuvant Therapy , Prognosis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Transcriptome , Trastuzumab/therapeutic use , Treatment Outcome , Gene Expression Profiling , Randomized Controlled Trials as Topic , Paclitaxel/therapeutic use
7.
J Med Chem ; 65(22): 15123-15139, 2022 11 24.
Article in English | MEDLINE | ID: mdl-36351049

ABSTRACT

To enhance the affinity of the human epidermal growth receptor 2 (HER2) targeted peptide developed previously, bispecific fusion peptides P1GCGT1 and P1GCGCGT1 were designed using an in silico approach. Molecular dynamic simulation showed that both peptides strongly interacted with HER2 domains II and IV. Compared with peptides targeting each single domain, P1GCGT1 and P1GCGCGT1 could bind to HER2 more significantly and targeted HER2-positive cells specifically. Additionally, both peptides were used to generate peptide-drug conjugates with camptothecin (CPT), among which I-1 and I-4 were screened for enhanced cellular activity and selectivity. Biological evaluation demonstrated that I-1 and I-4 induced cell apoptosis, promoted cell cycle arrestin S-phase, and inhibited Topo I activity. The binding affinity assay and confocal analysis revealed that I-1 and I-4 were effective at targeting HER2. Moreover, I-1 and I-4 showed better stability than single targeting peptide and presented enhanced antitumor activity and safety than CPT in tumor-bearing mice.


Subject(s)
Breast Neoplasms , Peptides , Receptor, ErbB-2 , Animals , Female , Humans , Mice , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Mice, Nude , Peptides/pharmacology , Peptides/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism
8.
Sci Rep ; 12(1): 10461, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35729189

ABSTRACT

HER-2 targeted therapies, such as monoclonal antibodies (mAbs) and CAR-T cell therapy have been applied in the treatment of various of cancers. However, the anti-HER2 CAR-T cell therapy are limited by its expensive production procedure and fatal side effects such as cytokine storm or "On target, off tumor". The application of anti-HER2 mAbs to the soild tumor are also plagued by the patients resistant with different mechanisms. Thus, the recombinant protein technology can be presented as an attractive methods in advantage its less toxic and lower cost. In this study, we produced a HER-2-targeting recombinant protein, which is the fusion of the anti-HER-2 single chain fragment variable domain, CCL19 and IL7 (HCI fusion protein). Our results showed that the recombinant protein can induce the specific lysis effects of immune cells on HER-2-positive gastric tumor cells and can suppress gastric tumor growth in a xenograft model by chemotactic autoimmune cell infiltration into tumor tissues and activated T cells. Taken together, our results revealed that the HCI fusion protein can be applied as a subsequent clinical drug in treating HER-2 positive gastric tumors.


Subject(s)
Chemokine CCL19 , Interleukin-7 , Receptors, Chimeric Antigen , Recombinant Fusion Proteins , Stomach Neoplasms , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Chemokine CCL19/genetics , Chemokine CCL19/pharmacology , Humans , Interleukin-7/genetics , Interleukin-7/pharmacology , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Xenograft Model Antitumor Assays
9.
Sci Rep ; 12(1): 3020, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194100

ABSTRACT

Radiopharmaceutical therapy (RPT) is an attractive strategy for treatment of disseminated cancers including those overexpressing the HER2 receptor including breast, ovarian and gastroesophageal carcinomas. Single-domain antibody fragments (sdAbs) exemplified by the HER2-targeted VHH_1028 evaluated herein are attractive for RPT because they rapidly accumulate in tumor and clear faster from normal tissues than intact antibodies. In this study, VHH_1028 was labeled using the residualizing prosthetic agent N-succinimidyl 3-guanidinomethyl 5-[131I]iodobenzoate (iso-[131I]SGMIB) and its tissue distribution evaluated in the HER2-expressing SKOV-3 ovarian and BT474 breast carcinoma xenograft models. In head-to-head comparisons to [131I]SGMIB-2Rs15d, a HER2-targeted radiopharmaceutical currently under clinical investigation, iso-[131I]SGMIB-VHH_1028 exhibited significantly higher tumor uptake and significantly lower kidney accumulation. The results demonstrated 2.9 and 6.3 times more favorable tumor-to-kidney radiation dose ratios in the SKOV-3 and BT474 xenograft models, respectively. Iso-[131I]SGMIB-VHH_1028 was prepared using a solid-phase extraction method for purification of the prosthetic agent intermediate Boc2-iso-[131I]SGMIB that reproducibly scaled to therapeutic-level doses and obviated the need for its HPLC purification. Single-dose (SKOV-3) and multiple-dose (BT474) treatment regimens demonstrated that iso-[131I]SGMIB-VHH_1028 was well tolerated and provided significant tumor growth delay and survival prolongation. This study suggests that iso-[131I]SGMIB-VHH_1028 is a promising candidate for RPT of HER2-expressing cancers and further development is warranted.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Gene Expression/genetics , Immunoglobulin Fragments/therapeutic use , Iodine Radioisotopes/pharmacology , Iodine Radioisotopes/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/therapeutic use , Animals , Disease Models, Animal , Female , Humans , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
10.
Front Immunol ; 13: 828386, 2022.
Article in English | MEDLINE | ID: mdl-35154149

ABSTRACT

Breast cancer has become the most commonly diagnosed cancer globally. The relapse and metastasis of breast cancer remain a great challenge despite advances in chemotherapy, endocrine therapy, and HER2 targeted therapy in the past decades. Innovative therapeutic strategies are still critically in need. Cancer vaccine is an attractive option as it aims to induce a durable immunologic response to eradicate tumor cells. Different types of breast cancer vaccines have been evaluated in clinical trials, but none has led to significant benefits. Despite the disappointing results at present, new promise from the latest study indicates the possibility of applying vaccines in combination with anti-HER2 monoclonal antibodies or immune checkpoint blockade. This review summarizes the principles and mechanisms underlying breast cancer vaccines, recapitulates the type and administration routes of vaccine, reviews the current results of relevant clinical trials, and addresses the potential reasons for the setbacks and future directions to explore.


Subject(s)
Breast Neoplasms/drug therapy , Cancer Vaccines/therapeutic use , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cancer Vaccines/classification , Cancer Vaccines/immunology , Drug Delivery Systems , Drug Therapy, Combination , Female , Humans , Immunotherapy/methods , Randomized Controlled Trials as Topic , Receptor, ErbB-2/immunology
11.
Protein Expr Purif ; 192: 106043, 2022 04.
Article in English | MEDLINE | ID: mdl-34973460

ABSTRACT

Antibody-drug conjugates (ADCs) are a major therapeutic tool for the treatment of advanced cancer. Malignant cells in advanced cancer often display multiple genetic mutations and become resistant to monotherapy. Therefore, a therapeutic regimen that simultaneously targets multiple molecules with multiple payloads is desirable. However, the development of ADCs is hampered by issues in biopharmaceutical manufacturing and the complexity of the conjugation process of low-molecular-weight payloads to biologicals. Here, we report antibody mimetic-drug conjugates (AMDCs) developed by exploiting the non-covalent binding property of payloads based on high-affinity binding of mutated streptavidin and modified iminobiotin. Miniprotein antibodies were fused to a low immunogenic streptavidin variant, which was then expressed in Escherichia coli inclusion bodies, solubilized, and refolded into functional tetramers. The AMDC developed against human epidermal growth factor receptor 2 (HER2) effectively killed cultured cancer cells using bis-iminobiotin conjugated to photo-activating silicon phthalocyanine. The HER2-targeting AMDC was also effective in vivo against a mouse KPL-4 xenograft model. This AMDC platform provides rapid, stable, and high-yield therapeutics against multiple targets.


Subject(s)
Escherichia coli/metabolism , Gene Expression , Immunoconjugates/genetics , Animals , Biotin/administration & dosage , Biotin/analogs & derivatives , Biotin/chemistry , Biotin/genetics , Biotin/immunology , Cell Line, Tumor , Cloning, Molecular , Escherichia coli/genetics , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Immunoconjugates/immunology , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Protein Folding , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Streptavidin/administration & dosage , Streptavidin/chemistry , Streptavidin/genetics , Streptavidin/immunology
12.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35086947

ABSTRACT

BACKGROUND: Tumorous heterogeneity is a hallmark of tumor evolution and cancer progression, being a longstanding challenge to targeted immunotherapy. Ex vivo armed T cells (EATs) using IgG-(L)-scFv bispecific antibodies (BsAbs) are potent tumor-specific cytotoxic effectors. To improve the anti-tumor efficacy of EATs against heterogeneous solid tumors, we explored multi-antigen targeting approaches. METHODS: Ex vivo expanded T cells were armed with BsAbs built on the IgG-(L)-scFv platform, where an anti-CD3 (huOKT3) scFv was attached to the carboxyl end of both light chains of a tumor specific IgG. Multispecificity was created by combining monospecific EATs, combining BsAbs on the same T cell, or combining specificities on the same antibody. Three multi-antigens targeting EAT strategies were tested: (1) pooled-EATs (EATs each with unique specificity administered simultaneously) or alternate-EATs (EATs each with unique specificity administered in an alternating schedule), (2) dual-EATs or multi-EATs (T cells simultaneously armed with ≥2 BsAbs), and (3) TriAb-EATs (T cells armed with BsAb specific for two targets besides CD3 (TriAb)). The properties and efficiencies of these three strategies were evaluated by flow cytometry, in vitro cytotoxicity, cytokine release assays, and in vivo studies performed in BALB-Rag2-/-IL-2R-γc-KO (BRG) mice xenografted with cancer cell line (CDX) or patient-derived tumor (PDX). RESULTS: Multi-EATs retained target antigen specificity and anti-tumor potency. Cytokine release with multi-EATs in the presence of tumor cells was substantially less than when multiple BsAbs were mixed with unarmed T cells. When tested against CDXs or PDXs, dual-EATs or multi-EATs effectively suppressed tumor growth without clinical toxicities. Most importantly, dual-EATs or multi-EATs were highly efficient in preventing clonal escape while mono-EATs or TriAb- EATs were not as effective. CONCLUSIONS: Multi-EATs have the potential to increase potency, reduce toxicity, and overcome tumor heterogeneity without excessive cytokine release. Arming T cells with multiple BsAbs deserves further exploration to prevent or to treat cancer resistance.


Subject(s)
Antibodies, Bispecific/therapeutic use , Gangliosides/immunology , Immunotherapy/methods , Neoplasms/therapy , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytokines/biosynthesis , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
Indian J Pathol Microbiol ; 65(1): 29-34, 2022.
Article in English | MEDLINE | ID: mdl-35074962

ABSTRACT

BACKGROUND: Gastric carcinoma is a major cause of cancer-related morbidity and mortality worldwide. Gastric neoplasms arise from genetic and epigenetic changes in various genes. Present study evaluates the immunoexpression of PTEN, HER2/neu, and Ki-67 in endoscopic gastric carcinoma biopsies and correlates the expression of these proteins with clinicopathological features. MATERIAL AND METHODS: Adequate endoscopic biopsies of 27 cases of gastric carcinoma were evaluated for World Health Organization (WHO) and Lauren's classification subtypes along with HER2/neu, PTEN, and Ki-67 immunoexpression. HER2/neu immunostaining was scored as proposed in the Trastuzumab for gastric cancer (ToGA) trial while PTEN staining and downregulation were assessed using an immunoreactive score. The cut-off for Ki-67 expression was taken as 90th percentile of the values in adjacent non-neoplastic tissue. All statistical analysis was done at 5% level of significance with SPSS v22 statistical software. RESULTS: Tubular adenocarcinoma was the commonest WHO histological subtype and 56% of cases were of intestinal type as per Lauren's classification. 55.6% of cases showed a complete loss of PTEN expression in neoplastic tissue. 17 of the 19 cases with adjacent non-neoplastic tissue showed PTEN downregulation in neoplastic tissue. 81.5% of cases had a high Ki-67 index and HER2/neu overexpression was noted in 36% of cases. All the four cases who died had high Ki-67 proliferation indices; 3 patients had loss of PTEN expression and HER2/neu overexpression. CONCLUSION: We conclude that these immunomarkers can play important role in the behavior of gastric carcinomas and can be targeted for new therapies.


Subject(s)
Gene Expression , Ki-67 Antigen/genetics , PTEN Phosphohydrolase/genetics , Receptor, ErbB-2/genetics , Stomach Neoplasms/classification , Stomach Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Aged , Aged, 80 and over , Biomarkers, Tumor/immunology , Biopsy , Endoscopy, Gastrointestinal/methods , Female , Gene Expression/immunology , Humans , Immunochemistry/methods , Ki-67 Antigen/immunology , Male , Middle Aged , PTEN Phosphohydrolase/immunology , Receptor, ErbB-2/immunology , Stomach/pathology , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Survival Analysis
14.
Front Immunol ; 13: 1042072, 2022.
Article in English | MEDLINE | ID: mdl-36591290

ABSTRACT

Objective: Besides breast and gastric cancer, HER2 amplification/mutation are also found in lung adenocarcinoma (LUAD). However, the correlation between HER2 variations and the phenotype of immunogenicity and tumor immune microenvironment (TIME) in LUAD compared with breast and gastric cancer has yet to be fully elucidated. Methods: We integrated public databases (discovery set) and internal data (validated set) of 288 patients representing three distinct HER2-altered tumors. Genomic data were used to identify somatic mutations, copy number variations, and calculate tumor mutational burden (TMB) and microsatellite instability score. RNA sequencing was conducted to estimate immune gene signatures and contents of tumor-infiltrating immune cell populations. Finally, IHC was used to determine PD-L1 expression and the tumoral-infiltration of immune cells in 50 HER2-variant tumor specimens with no prior therapeutic regimens. Results: Compared with HER2-amplified breast and gastric cancers, patients with HER2-amplified LUAD showed higher immunogenicity, mainly manifested in immune checkpoints expression and tissue/blood TMB. Additionally, HER2-amplified LUAD exhibited an inflamed TIME with remarkably increased genes encoding HLAs, T-cell activity and immune cell-type, and accompanied with tumor-infiltrating lymphocytes. In LUAD, patients with HER2 amplification possessed higher tissue TMB than HER2 mutation, whereas no difference was observed in PD-L1 expression. HER2 amplification (primary) was associated with significantly higher PD-L1 expression and TMB than acquired HER2 amplification after resistance to EGFR-TKIs. Conclusion: Patients with HER2-amplified LUAD have better immunogenicity and/or an inflamed TIME among HER2-aberrant tumors. Our study may provide clues for establishing the benefits and uses of ICIs for patients with this disease.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Receptor, ErbB-2 , Stomach Neoplasms , Tumor Microenvironment , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , DNA Copy Number Variations , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology
15.
J Mol Biol ; 434(8): 167412, 2022 04 30.
Article in English | MEDLINE | ID: mdl-34942167

ABSTRACT

While 20 canonical amino acids are used by most organisms for protein synthesis, the creation of cells that can use noncanonical amino acids (ncAAs) as additional protein building blocks holds great promise for preparing novel medicines and for studying complex questions in biological systems. However, only a small number of biosynthetic pathways for ncAAs have been reported to date, greatly restricting our ability to generate cells with ncAA building blocks. In this study, we report the creation of a completely autonomous bacterium that utilizes 3,4-dihydroxy-L-phenylalanine (DOPA) as its 21st amino acid building block. Like canonical amino acids, DOPA can be biosynthesized without exogenous addition and can be genetically incorporated into proteins in a site-specific manner. Equally important, the protein production yields of DOPA-containing proteins from these autonomous cells are greater than those from cells exogenously fed with 9 mM DOPA. The unique catechol moiety of DOPA can be used as a versatile handle for site-specific protein functionalizations via either oxidative coupling or strain-promoted oxidation-controlled cyclooctyne-1,2-quinone (SPOCQ) cycloaddition reactions. We further demonstrate the use of these autonomous cells in preparing fluorophore-labeled anti-human epidermal growth factor 2 (HER2) antibodies for the detection of HER2 expression on cancer cells.


Subject(s)
Cell Engineering , Dihydroxyphenylalanine , Escherichia coli , Protein Biosynthesis , Antibodies , Breast Neoplasms/metabolism , Dihydroxyphenylalanine/genetics , Dihydroxyphenylalanine/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genetic Code , Humans , Receptor, ErbB-2/analysis , Receptor, ErbB-2/immunology
16.
Int J Cancer ; 150(1): 164-173, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34480368

ABSTRACT

Checkpoint-blockade therapy (CBT) is approved for select colorectal cancer (CRC) patents, but additional immunotherapeutic options are needed. We hypothesized that vaccination with carcinoembryonic antigen (CEA) and Her2/neu (Her2) peptides would be immunogenic and well tolerated by participants with advanced CRC. A pilot clinical trial (NCT00091286) was conducted in HLA-A2+ or -A3+ Stage IIIC-IV CRC patients. Participants were vaccinated weekly with CEA and Her2 peptides plus tetanus peptide and GM-CSF emulsified in Montanide ISA-51 adjuvant for 3 weeks. Adverse events (AEs) were recorded per NIH Common Terminology Criteria for Adverse Events version 3. Immunogenicity was evaluated by interferon-gamma ELISpot assay of in vitro sensitized peripheral blood mononuclear cells and lymphocytes from the sentinel immunized node. Eleven participants were enrolled and treated; one was retrospectively found to be ineligible due to HLA type. All 11 participants were included in AEs and survival analyses, and the 10 eligible participants were evaluated for immunogenicity. All participants reported AEs: 82% were Grade 1-2, most commonly fatigue or injection site reactions. Two participants (18%) experienced treatment-related dose-limiting Grade 3 AEs; both were self-limiting. Immune responses to Her2 or CEA peptides were detected in 70% of participants. Median overall survival (OS) was 16 months; among those enrolled with no evidence of disease (n = 3), median OS was not reached after 10 years of follow-up. These data demonstrate that vaccination with CEA or Her2 peptides is well tolerated and immunogenic. Further study is warranted to assess potential clinical benefits of vaccination in advanced CRC either alone or in combination with CBT.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/drug therapy , Dendritic Cells/immunology , Peptide Fragments/therapeutic use , Receptor, ErbB-2/immunology , Vaccination/methods , Adult , Aged , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , GPI-Linked Proteins/immunology , Humans , Male , Middle Aged , Peptide Fragments/immunology , Pilot Projects , Prognosis , Retrospective Studies , Survival Rate
17.
Protein Eng Des Sel ; 342021 02 15.
Article in English | MEDLINE | ID: mdl-34908139

ABSTRACT

Human epidermal growth factor receptor 2 (HER2) protein overexpression is found in ~30% of invasive breast carcinomas and in a high proportion of noninvasive ductal carcinomas in situ. Targeted cancer therapy is based on monoclonal antibodies and kinase inhibitors and reflects a new era of cancer therapy. However, delivery to tumor cells in vivo is hampered by the large size (150 kDa) of conventional antibodies. Furthermore, there are many disadvantages with the current anti-HER2 drug, including drug resistance and adverse effects. Nanobodies (15 kDa), single-domain antibody (sdAb) fragments, can overcome these limitations. This study produced the recombinant sdAb against the HER2-tyrosine kinase (HER2-TK) domain using phage display technology. Three specific anti-HER2-TK sdAbs were selected for further characterization. Hallmark VHH residue identification and amino acid sequence analysis revealed that clone numbers 4 and 22 were VH antibodies, whereas clone number 17 was a VH H antibody (nanobody). The half-maximal inhibitory concentration of VHH17 exhibited significantly greater HER2 kinase-inhibition activity than the other clones. Consistent with these results, several charges and polar residues of the HER2-TK activation loop that were predicted based on mimotope analysis also appeared in the docking result and interacted via the CDR1, CDR2 and CDR3 loops of VHH17. Furthermore, the cell-penetrable VHH17 (R9 VHH17) showed cell-penetrability and significantly decreased HER2-positive cancer cell viability. Thus, the VH H17 could be developed as an effective therapeutic agent to treat HER2-positive breast cancer.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2/immunology , Single-Domain Antibodies , Antibodies, Monoclonal , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Surface Display Techniques , Early Detection of Cancer , Female , Humans , Single-Domain Antibodies/genetics
18.
Int Immunopharmacol ; 101(Pt B): 108368, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34857479

ABSTRACT

Due to the high affinity for binding to target molecules and also other unique attributes, affibodies have a great potential to be used in immunotherapeutic and diagnostic approaches. However, the possibility of undesirable immune response is still a great concern. In the current study, we investigated the possible antigenicity, allergenicity and cytotoxicity of the HER2-targeting affibody ZHER2. The binding affinity of potential epitopes of the affibody to murine major histocompatibility complex (MHC) molecules was investigated by immunoinformatics tools and docking approaches. The possible interaction of ZHER2 with human leukocyte antigens HLA-DP, HLA-DM, HLA-DQ and HLA-DR was also studied by protein-protein docking. Additionally, the synthesized affibody gene was expressed and the protein was purified for boosted immunization of Balb/c mice. Induced secretion of IFN-γ, IL-2, IL-4 and IL-10, and total serum IgG were assessed in the immunized mice. Furthermore, MTT cell viability test was performed to evaluate the cytotoxicity of ZHER2 in splenocytes of the treated mice. In silico analyses showed the possible induction of the immune response by ZHER2. While the affibody could elicit the secretion of cellular immune cytokines, it could not induce a significant humoral response in the treated mice and did not show any cytotoxic effects on the exposed splenocytes. These findings explain the practicability of ZHER2 for therapeutic and in vivo diagnostic usages, though its ubiquitous application may need more studies.


Subject(s)
Antibodies , Molecular Mimicry , Receptor, ErbB-2/immunology , Recombinant Fusion Proteins/immunology , Allergens , Animals , Cell Survival , Cloning, Molecular , Computer Simulation , Cytokines/genetics , Cytokines/metabolism , Epitopes , Female , Gene Expression Regulation/drug effects , Mice , Mice, Inbred BALB C , Models, Molecular , Protein Conformation , Protein Domains
19.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34830027

ABSTRACT

Breast cancer (BC) is a heterogeneous disease composed of multiple subtypes with different molecular characteristics and clinical outcomes. The metastatic process in BC depends on the transcription factors (TFs) related to epithelial-mesenchymal transition (EMT), including the master regulator Twist1. However, its role beyond EMT in BC subtypes remains unclear. Our study aimed to investigate the role of Twist1, beyond EMT, in the molecular subtypes of BC. In patients, we observed the overexpression of TWIST1 in the HER2+ group. The silencing of TWIST1 in HER2+ BC cells resulted in the upregulation of 138 genes and the downregulation of 174 genes compared to control cells in a microarray assay. In silico analysis revealed correlations between Twist1 and important biological processes such as the Th17-mediated immune response, suggesting that Twist1 could be relevant for IL-17 signaling in HER2+ BC. IL-17 signaling was then examined, and it was shown that TWIST1 knockdown caused the downregulation of leading members of IL-17 signaling pathway. Taken together, our findings suggest that Twist1 plays a role on IL-17 signaling in HER2+ BC.


Subject(s)
Breast Neoplasms/immunology , Gene Expression Regulation, Neoplastic/immunology , Interleukin-17/immunology , Nuclear Proteins/immunology , Receptor, ErbB-2/immunology , Signal Transduction/immunology , Twist-Related Protein 1/immunology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Interleukin-17/genetics , Nuclear Proteins/genetics , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Twist-Related Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...