Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Ther ; 32(9): 3114-3127, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-38910324

ABSTRACT

T cell-focused cancer immunotherapy including checkpoint inhibitors and cell therapies has been rapidly evolving over the past decade. Nevertheless, there remains a major unmet medical need in oncology generally and immuno-oncology specifically. We have constructed an oncolytic adenovirus, Ad5/3-E2F-d24-aMUC1aCD3-IL-2 (TILT-322), which is armed with a human aMUC1aCD3 T cell engager and IL-2. TILT-322 treatment stimulated T cell cytotoxicity through the increased presence of granzyme B, perforin, and interferon-gamma. Additional immune profiling indicated TILT-322 increased gamma delta T cell activation and impacted other cell types such as natural killer cells and natural killer-like T cells that are traditionally involved in cancer immunotherapy. TILT-322 treatment also decreased the proportion of exhausted CD8+ T cells as demarked by immune checkpoint expression in ovarian ascites samples. Overall, our data showed that TILT-322 treatment led to an enhanced T cell activation and reversed T cell exhaustion translating into high antitumor efficacy when given locally or intravenously. The analysis of blood and tumors isolated from an in vivo patient-derived ovarian cancer xenograft model suggested TILT-322 mediated tumor control through improved T cell functions. Therefore, TILT-322 is a promising novel anti-tumor agent for clinical translation.


Subject(s)
Adenoviridae , Antibodies, Bispecific , Ascites , Interleukin-2 , Mucin-1 , Ovarian Neoplasms , Xenograft Model Antitumor Assays , Humans , Ovarian Neoplasms/therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/genetics , Female , Animals , Adenoviridae/genetics , Mice , Cell Line, Tumor , Ascites/therapy , Ascites/immunology , Interleukin-2/metabolism , Mucin-1/genetics , Mucin-1/immunology , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Disease Models, Animal , Immunotherapy/methods , T-Cell Exhaustion
2.
J Biol Chem ; 299(7): 104883, 2023 07.
Article in English | MEDLINE | ID: mdl-37269947

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy has had considerable success in the treatment of B-cell malignancies. Targeting the B-lineage marker CD19 has brought great advances to the treatment of acute lymphoblastic leukemia and B-cell lymphomas. However, relapse remains an issue in many cases. Such relapse can result from downregulation or loss of CD19 from the malignant cell population or expression of alternate isoforms. Consequently, there remains a need to target alternative B-cell antigens and diversify the spectrum of epitopes targeted within the same antigen. CD22 has been identified as a substitute target in cases of CD19-negative relapse. One anti-CD22 antibody-clone m971-targets a membrane-proximal epitope of CD22 and has been widely validated and used in the clinic. Here, we have compared m971-CAR with a novel CAR derived from IS7, an antibody that targets a central epitope on CD22. The IS7-CAR has superior avidity and is active and specific against CD22-positive targets, including B-acute lymphoblastic leukemia patient-derived xenograft samples. Side-by-side comparisons indicated that while IS7-CAR killed less rapidly than m971-CAR in vitro, it remains efficient in controlling lymphoma xenograft models in vivo. Thus, IS7-CAR presents a potential alternative candidate for the treatment of refractory B-cell malignancies.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Chimeric Antigen , Sialic Acid Binding Ig-like Lectin 2 , Humans , Antigens, CD19 , Epitopes , Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Recurrence
3.
Br J Haematol ; 204(2): 389-390, 2024 02.
Article in English | MEDLINE | ID: mdl-37964473

ABSTRACT

Personalized treatment options for subsets of patients with DLBCL are beginning to emerge. Caracciolo et al. explore UMG1, an epitope of CD43 as a potential target for certain patients with DLBCL, and demonstrate promising preclinical activity of an Anti-UMG1-antibody. Commentary on: Caracciolo et al. UMG1/CD3ε-bispecific T-cell engager (BTCE) redirects T-cell cytotoxicity against diffuse large B-cell lymphoma (DLBCL). Br J Haematol 2024;204:555-560.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Immunotherapy , T-Lymphocytes
4.
Br J Haematol ; 204(2): 555-560, 2024 02.
Article in English | MEDLINE | ID: mdl-37963444

ABSTRACT

UMG1 is a unique epitope of CD43, not expressed by normal cells and tissues of haematopoietic and non-haematopoietic origin, except thymocytes and a minority (<5%) of peripheral blood T lymphocytes. By immunohistochemistry analysis of tissue microarray and pathology slides, we found high UMG1 expression in 20%-24% of diffuse large B-cell lymphomas (DLBCLs), including highly aggressive BCL2high and CD20low cases. UMG1 membrane expression was also found in DLBCL bone marrow-infiltrating cells and established cell lines. Targeting UMG1 with a novel asymmetric UMG1/CD3ε-bispecific T-cell engager (BTCE) induced redirected cytotoxicity against DLBCL cells and was synergistic with lenalidomide. We conclude that UMG1/CD3ε-BTCE is a promising therapeutic for DLBCLs.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , T-Lymphocytes , Humans , T-Lymphocytes/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Immunohistochemistry
5.
Cancer Immunol Immunother ; 73(10): 196, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105814

ABSTRACT

Anti-cluster of differentiation (CD) 3 × α programmed death-ligand 1 (PD-L1) bispecific T-cell engager (BsTE)-bound T-cells (BsTE:T) are a promising new cancer treatment agent. However, the mechanisms of action of bispecific antibody-armed activated T-cells are poorly understood. Therefore, this study aimed to investigate the anti-tumor mechanism and efficacy of BsTE:T. The BsTE:T migration was assessed in vivo and in vitro using syngeneic and xenogeneic tumor models, flow cytometry, immunofluorescence staining, transwell migration assays, microfluidic chips, Exo View R100, western blotting, and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology. In murine B16 melanoma, MC38 colon cancer, and human multiple myeloma cells, BsTE:T exhibited superior tumor elimination relative to that of T-cells or BsTE alone. Moreover, BsTE:T migration into tumors was significantly enhanced owing to the presence of PD-L1 in tumor cells and secretion of PD-L1-containing exosomes. Furthermore, increased infiltration of CD44highCD62Llow effector memory CD8+ T-cells into tumors was closely associated with the anti-tumor effect of BsTE:T. Therefore, BsTE:T is an innovative potential anti-tumor therapy, and exosomal PD-L1 plays a crucial role both in vitro and in vivo in the anti-tumor activity of BsTE:T.


Subject(s)
Antibodies, Bispecific , B7-H1 Antigen , Exosomes , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/immunology , Exosomes/metabolism , Exosomes/immunology , Mice , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice, Inbred C57BL , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , CD3 Complex/immunology , CD3 Complex/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Cell Line, Tumor , Female , Cell Movement , Xenograft Model Antitumor Assays
6.
Immunol Rev ; 298(1): 84-98, 2020 11.
Article in English | MEDLINE | ID: mdl-33048357

ABSTRACT

Human γδ T lymphocytes are predominated by two major subsets, defined by the variable domain of the δ chain. Both, Vδ1 and Vδ2 T cells infiltrate in tumors and have been implicated in cancer immunosurveillance. Since the localization and distribution of tumor-infiltrating γδ T cell subsets and their impact on survival of cancer patients are not completely defined, this review summarizes the current knowledge about this issue. Different intrinsic tumor resistance mechanisms and immunosuppressive molecules of immune cells in the tumor microenvironment have been reported to negatively influence functional properties of γδ T cell subsets. Here, we focus on selected tumor resistance mechanisms including overexpression of cyclooxygenase (COX)-2 and indolamine-2,3-dioxygenase (IDO)-1/2, regulation by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/TRAIL-R4 pathway and the release of galectins. These inhibitory mechanisms play important roles in the cross-talk of γδ T cell subsets and tumor cells, thereby influencing cytotoxicity or proliferation of γδ T cells and limiting a successful γδ T cell-based immunotherapy. Possible future directions of a combined therapy of adoptively transferred γδ T cells together with γδ-targeting bispecific T cell engagers and COX-2 or IDO-1/2 inhibitors or targeting sialoglycan-Siglec pathways will be discussed and considered as attractive therapeutic options to overcome the immunosuppressive tumor microenvironment.


Subject(s)
Neoplasms , Receptors, Antigen, T-Cell, gamma-delta , Humans , Lymphocyte Activation , Neoplasms/therapy , T-Lymphocyte Subsets , Tumor Microenvironment
7.
Clin Immunol ; 255: 109755, 2023 10.
Article in English | MEDLINE | ID: mdl-37673224

ABSTRACT

Severe eosinophilic asthma (SEA) is characterized by elevated eosinophil counts in the blood and airway mucosa. While monoclonal antibody therapies targeting interleukin-5 (IL-5) and its receptor (IL-5Rα) have improved treatment, some patients remain unresponsive. We propose an alternative approach to eliminate eosinophils using T cells by engineering IL-5Rα × CD3 bispecific T-cell engagers (bsTCEs) that target both IL-5Rα on eosinophils and CD3 on T cells. We designed different formats of IL-5Rα × CD3 bsTCEs, incorporating variations in valency, geometry, and affinity for the target antigen binding. We identified the single-chain variable fragment (scFv)-Fc format with the highest affinity toward the membrane-proximal domain of IL-5Rα in the IL-5Rα-binding arm showed the most potent cytotoxicity against IL-5Rα-expressing peripheral eosinophils by activating autologous primary T cells from healthy donors. This study proposes IL-5Rα × CD3 bsTCEs as potential alternatives for SEA treatment. Importantly, it demonstrates the first application of bsTCEs in eliminating disease-associated cells, including eosinophils, beyond cancer cells.


Subject(s)
Asthma , Eosinophils , Humans , T-Lymphocytes/metabolism , Antibodies, Monoclonal/metabolism
8.
Br J Haematol ; 201(3): 417-421, 2023 05.
Article in English | MEDLINE | ID: mdl-35594370

ABSTRACT

The delivery of bispecific antibodies (BsAbs) targeting B-cell maturation antigen (BCMA) and CD3 using the gene therapy approach is a promising alternative for BsAb administration in patients with multiple myeloma (MM). In the present study, we evaluated the efficacy of this approach using a xenograft model. Tumour growth was significantly delayed in mice treated with single electroporation-enhanced intramuscular injection of plasmid DNA encoding BCMA/CD3 BsAb in contrast to the vehicle control-treated group. Limited toxicity was observed following treatment. This study demonstrates that the gene therapy-based approach for the delivery of BCMA/CD3 BsAb is effective and safe for the treatment of MM.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Humans , Mice , Animals , Multiple Myeloma/genetics , Multiple Myeloma/therapy , B-Cell Maturation Antigen/genetics , T-Lymphocytes , CD3 Complex , Antibodies, Bispecific/therapeutic use , Plasmids/genetics
9.
Cancer Immunol Immunother ; 72(9): 2939-2948, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37247022

ABSTRACT

Immunotherapy has revolutionized cancer therapy. Two recently FDA-approved immunotherapies for B-cell malignancies target CD19, in the form of a Bispecific T-Cell Engager (BiTE) antibody construct or chimeric antigen receptor T (CAR-T) cells. Blinatumomab, an FDA-approved BiTE, binds to CD19 on B cells and to CD3 on T cells, mediating effector-target cell contact and T-cell activation that results in effective elimination of target B cells. Although CD19 is expressed by essentially all B-cell malignancies at clinical presentation, relapses with loss or reduction in CD19 surface expression are increasingly recognized as a cause of treatment failure. Therefore, there is a clear need to develop therapeutics for alternate targets. We have developed a novel BiTE consisting of humanized anti-CD22 and anti-CD3 single chain variable fragments. Target binding of the anti-CD22 and anti-CD3 moieties was confirmed by flow cytometry. CD22-BiTE promoted in vitro cell-mediated cytotoxicity in a dose and effector: target (E:T)-dependent fashion. Additionally, in an established acute lymphoblastic leukemia (ALL) xenograft mouse model, CD22-BiTE demonstrated tumor growth inhibition, comparable to blinatumomab. Further, the combination of blinatumomab and CD22-BiTE yielded increased efficacy in vivo when compared to the single agents. In conclusion, we report here the development of a new BiTE with cytotoxic activity against CD22+ cells which could represent an alternate or complementary therapeutic option for B-cell malignancies.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Animals , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Antineoplastic Agents/therapeutic use , T-Lymphocytes , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antigens, CD19
10.
J Transl Med ; 21(1): 301, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37143061

ABSTRACT

BACKGROUND: Pronectins™ are a new class of fibronectin-3-domain 14th-derived (14Fn3) antibody mimics that can be engineered as bispecific T cell engager (BTCE) to redirect immune effector cells against cancer. We describe here the in vitro and in vivo activity of a Pronectin™ AXL-targeted first-in-class bispecific T cell engager (pAXLxCD3ε) against Epithelial Ovarian Cancer (EOC). METHODS: pAXLxCD3ε T-cell mediated cytotoxicity was evaluated by flow cytometry and bioluminescence. pAXLxCD3ε mediated T-cell infiltration, activation and proliferation were assessed by immunofluorescence microscopy and by flow cytometry. Activity of pAXLxCD3ε was also investigated in combination with poly-ADP ribose polymerase inhibitors (PARPi). In vivo antitumor activity of pAXLxCD3ε was evaluated in immunocompromised (NSG) mice bearing intraperitoneal or subcutaneous EOC xenografts and immunologically reconstituted with human peripheral blood mononuclear cells (PBMC). RESULTS: pAXLxCD3ε induced dose-dependent cytotoxicity by activation of T lymphocytes against EOC cells, regardless of their histologic origin. The addition of PARPi to cell cultures enhanced pAXLxCD3ε cytotoxicity. Importantly, in vivo, pAXLxCD3ε was highly effective against EOC xenografts in two different NSG mouse models, by inhibiting the growth of tumor cells in ascites and subcutaneous xenografts. This effect translated into a significantly prolonged survival of treated animals. CONCLUSION: pAXLxCD3ε is an active therapeutics against EOC cells providing a rational for its development as a novel agent in this still incurable disease. The preclinical validation of a first-in-class agent opens the way to the development of a new 14Fn3-based scaffold platform for the generation of innovative immune therapeutics against cancer.


Subject(s)
Antibodies, Bispecific , Ovarian Neoplasms , Humans , Mice , Animals , Female , Leukocytes, Mononuclear , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Ovarian Neoplasms/drug therapy , T-Lymphocytes , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , CD3 Complex
11.
Cell Immunol ; 393-394: 104787, 2023.
Article in English | MEDLINE | ID: mdl-37976975

ABSTRACT

The administration of blinatumomab was accompanied by several adverse effects, including activation of regulatory T-cells and cytokine storm. The objective of this study was to produce and evaluate a novel αCD8/CD19 BiTE (αCD8/CD19) with the potency to directly target CD8+T-cells. In-silico studies were utilized for determining proper folding, receptor binding, and structural stability of αCD8/CD19 protein. Western blotting and indirect surface staining were used to evaluate the size accuracy and binding potency of the purified protein. Functionality was assessed for granzyme B production, cytotoxicity, and proliferation. TheαCD8/CD19recombinant protein was produced in the CHO-K1 cell line with a final concentration of 1.94 mg/l. The αCD8/CD19 bound to CD8+and CD19+cell lines and induced significant granzyme B production, cytotoxic activity and proliferation potential in the presence of IL-2 and tumor target cells. The maximum CD8+T-cell biological activity was observed on the 10th day with 10:1 effector-to-target ratio.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Neoplasms , Humans , Granzymes , Neoplasms/pathology , CD8-Positive T-Lymphocytes/metabolism , Antineoplastic Agents/pharmacology , Antibodies, Bispecific/adverse effects , Antigens, CD19
12.
Mol Pharm ; 20(5): 2754-2760, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37067377

ABSTRACT

The elevated expression of GPNMB and VCAM-1 has been observed in many cancers including breast cancer, melanoma, and prostate cancers. Such overexpression of GPNMB and VCAM-1 has been associated with poor prognosis and increased cancer metastasis. Thus, GPNMB and VCAM-1 are potential targets for immunotherapies across multiple cancers. In this study, two high-affinity specific human VH domain antibody candidates, 87 (GPNMB) and 1B2 (VCAM-1), were isolated from our in-house proprietary phage-displayed human VH antibody domain libraries. The avidity was increased after conversion to VH-Fc. Domain-based bispecific T-cell engagers (DbTE) based on these two antibodies combined with the anti-CD3ε OKT3 antibody exhibited potent killing against GPNMB and VCAM-1-positive cancer cells, respectively. Hence, these two domain antibodies are promising therapeutic candidates for cancers expressing GPNMB or VCAM-1.


Subject(s)
Breast Neoplasms , Melanoma , Humans , Female , Vascular Cell Adhesion Molecule-1 , Antibodies , Breast Neoplasms/drug therapy , Immunoglobulin Variable Region , Transcription Factors , Membrane Glycoproteins
13.
Pediatr Blood Cancer ; : e30435, 2023 May 28.
Article in English | MEDLINE | ID: mdl-37246348

ABSTRACT

We analyzed changes in laboratory parameters, including blood counts, liver enzymes, inflammation and coagulation markers, and cytokines, from 70 blinatumomab-treated pediatric patients (NCT01471782). Overall, trends were consistent in responders and nonresponders. Platelets and lymphocytes peaked on day (D) 10 in cycle 1 and returned to baseline on D42 and D29, respectively. Neutrophils peaked on D2 and returned to baseline on D42. Alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and bilirubin peaked on D17, reversing to baseline on D29; total protein levels were constant. These findings indicate that blinatumomab-induced changes in laboratory parameters were transient, reversible, and not requiring treatment interruptions in responders and nonresponders.

14.
Mol Ther ; 30(7): 2537-2553, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35570396

ABSTRACT

Bispecific T cell engagers (BiTEs) are bispecific antibodies that redirect T cells to target antigen-expressing tumors. We hypothesized that BiTE-secreting T cells could be a valuable therapy in solid tumors, with distinct properties in mono- or multi-valent strategies incorporating chimeric antigen receptor (CAR) T cells. Glioblastomas represent a good model for solid tumor heterogeneity, representing a significant therapeutic challenge. We detected expression of tumor-associated epidermal growth factor receptor (EGFR), EGFR variant III, and interleukin-13 receptor alpha 2 (IL13Rα2) on glioma tissues and cancer stem cells. These antigens formed the basis of a multivalent approach, using a conformation-specific tumor-related EGFR targeting antibody (806) and Hu08, an IL13Rα2-targeting antibody, as the single chain variable fragments to generate new BiTE molecules. Compared with CAR T cells, BiTE T cells demonstrated prominent activation, cytokine production, and cytotoxicity in response to target-positive gliomas. Superior response activity was also demonstrated in BiTE-secreting bivalent T cells compared with bivalent CAR T cells in a glioma mouse model at early phase, but not in the long term. In summary, BiTEs secreted by mono- or multi-valent T cells have potent anti-tumor activity in vitro and in vivo with significant sensitivity and specificity, demonstrating a promising strategy in solid tumor therapy.


Subject(s)
Glioblastoma , Interleukin-13 Receptor alpha2 Subunit , Animals , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/pathology , Immunotherapy, Adoptive , Mice , T-Lymphocytes , Xenograft Model Antitumor Assays
15.
Acta Haematol ; 145(6): 592-602, 2022.
Article in English | MEDLINE | ID: mdl-35790143

ABSTRACT

INTRODUCTION: The safety and efficacy of blinatumomab, a CD19/CD3 bispecific T-cell engager (BiTE®) molecule, was evaluated in an expansion cohort of the phase 1b/2 study (NCT02412306) in Japanese adult (n = 14) and pediatric (n = 17) patients with relapsed/refractory Philadelphia-negative B-cell precursor (BCP) acute lymphoblastic leukemia (ALL). MATERIALS AND METHODS: Globally recommended blinatumomab doses were administered to adult (9-28 µg/day) and pediatric (5-15 µg/m2/day) patients. Primary endpoint was the incidence of treatment-emergent adverse events (TEAEs) and treatment-related AEs. RESULTS: All adult and pediatric patients experienced ≥1 TEAE. Grade ≥3 TEAEs were observed in 11 (79%) adult and 15 (88%) pediatric patients. Blinatumomab was discontinued in 1 (6%) pediatric patient due to treatment-related grade 4 cytokine release syndrome. Fatal AEs such as disease progression and multiple-organ dysfunction syndrome, which were not treatment-related, were reported in 2 (12%) pediatric patients. Eleven (79%) adults achieved complete remission (CR)/CR with partial hematological recovery (CRh) within the first two blinatumomab cycles. Nine of 10 adult patients with CR/CRh and evaluable minimal residual disease (MRD) achieved MRD response. CR/CRh was achieved by 5 (29%) pediatric patients, of which two had MRD response. CONCLUSION: In conclusion, blinatumomab was safe and efficacious in Japanese patients with relapsed/refractory BCP ALL.


Subject(s)
Antineoplastic Agents , Lymphoma, B-Cell , Neoplasm Recurrence, Local , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Adult , Child , Humans , Acute Disease , Antineoplastic Agents/adverse effects , Japan , Lymphoma, B-Cell/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Neoplasm Recurrence, Local/drug therapy
16.
Curr Treat Options Oncol ; 23(2): 155-170, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35182296

ABSTRACT

OPINION STATEMENT: While there have been numerous advances in the field of non-Hodgkin lymphoma (NHL) over the last decade, relapsed and/or refractory (R/R) NHL remains a challenge and an area with unmet needs. T-cell redirecting immunotherapeutic approaches including chimeric antigen receptor (CAR) T-cells and bispecific antibodies (BsAbs) have the potential to revolutionize NHL therapy. BsAbs target CD3 on T-cells and CD19 or CD20 on malignant B-cells and have shown promises as a novel immunotherapy for NHL. The development of CD19 × CD3 BsAbs such as blinatumomab was met with significant challenges due to dose-limiting neurologic side effects. However, several CD20 × CD3 BsAbs including odronextamab, mosunetuzumab, glofitamab, and epcoritamab emerged recently. They have favorable toxicity profiles, with reduced cytokine release syndrome and neurotoxicity. In addition, all these BsAbs have demonstrated very promising efficacy in R/R NHL. With expansion and registrational studies actively ongoing, approvals of these agents for R/R NHL are anticipated in the near future. Some important questions pertinent to future clinical development of BsAbs include when and how to best utilize BsAbs in the management of R/R NHL, whether there is a role of BsAbs in treatment-naïve NHL, and how to combine BsAbs with other therapies. For example, whether BsAbs can be combined with cytotoxic chemotherapy effectively remains to be seen. A plethora of clinical studies will be needed to help address these questions, some of which are already ongoing. In addition, how do BsAbs compare to CAR T-cell therapy and how to choose and sequence between BsAbs and CAR T-cell therapy need to be addressed. While many of these critical questions remain to be answered in clinical studies, we believe the future of BsAbs in the NHL is very bright.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Lymphoma, Non-Hodgkin , Antibodies, Bispecific/therapeutic use , Antigens, CD19 , Antineoplastic Agents/therapeutic use , Humans , Immunotherapy, Adoptive/adverse effects , Lymphoma, Non-Hodgkin/drug therapy , T-Lymphocytes
17.
Biol Pharm Bull ; 45(4): 429-437, 2022.
Article in English | MEDLINE | ID: mdl-35370267

ABSTRACT

Cancer immunotherapies are powerful therapeutic options for cancer patients. To enhance the therapeutic effects of cancer immunotherapies, we plan to develop novel immunostimulatory drugs for use in combination with cancer immunotherapy. In the present study, we focused on tetracyclines, the effects of which are controversial for immunotherapy. We examined the effects of tetracyclines on human T cells in the peripheral blood of healthy donors and the tumor tissues of non-small cell lung cancer (NSCLC) patients. By using bispecific T-cell engager technology to assess the cytotoxicity of peripheral T cells against tumor cells, we showed that tetracyclines (minocycline, tetracycline, doxycycline, meclocycline, chlortetracycline, and demeclocycline) enhanced T-cell cytotoxicity through granzyme B expression and CD4+ and CD8+ T-cell proliferation. In analyses of the peripheral blood mononuclear cells (PBMCs) and lung tumor-infiltrated cells of NSCLC patients, we found that demeclocycline enhanced T-cell cytotoxicity not only in PBMCs, but also in lung tumor tissues. These results support the further application of tetracyclines to combination cancer immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Leukocytes, Mononuclear , Lung Neoplasms/drug therapy , Minocycline , T-Lymphocytes
18.
Int J Mol Sci ; 23(14)2022 Jul 18.
Article in English | MEDLINE | ID: mdl-35887271

ABSTRACT

Radiation of tumor cells can lead to the selection and outgrowth of tumor escape variants. As radioresistant tumor cells are still sensitive to retargeting of T cells, it appears promising to combine radio- with immunotherapy keeping in mind that the radiation of tumors favors the local conditions for immunotherapy. However, radiation of solid tumors will not only hit the tumor cells but also the infiltrated immune cells. Therefore, we wanted to learn how radiation influences the functionality of T cells with respect to retargeting to tumor cells via a conventional bispecific T cell engager (BiTE) and our previously described modular BiTE format UNImAb. T cells were irradiated between 2 and 50 Gy. Low dose radiation of T cells up to about 20 Gy caused an increased release of the cytokines IL-2, TNF and interferon-γ and an improved capability to kill target cells. Although radiation with 50 Gy strongly reduced the function of the T cells, it did not completely abrogate the functionality of the T cells.


Subject(s)
Antibodies, Bispecific , Prostatic Neoplasms , Humans , Immunologic Factors , Immunotherapy/methods , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , T-Lymphocytes
19.
Int J Mol Sci ; 23(23)2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36498915

ABSTRACT

The growing understanding of the molecular mechanisms of carcinogenesis accelerated the development of monoclonal therapeutic antibodies to specifically target multiple cancer pathways. Recombinant protein therapeutics now constitute a large proportion of yearly approved medicines. Oncology, autoimmune diseases and to a smaller degree the prophylaxis of organ transplant rejection are their main application areas. As of the date of this review, 37 monoclonal antibody products are approved for use in cancer treatments in the United Kingdom. Currently, the antibody therapeutics market is dominated by monoclonal immunoglobulins (IgGs). New types of recombinant antibody therapeutics developed more recently include bispecific recombinant antibodies and other recombinantly produced functional proteins. This review focuses on the approved therapeutic antibodies used in cancer treatment in the UK today and describes their antigen targets and molecular mechanisms involved. We provide convenient links to the relevant databases and other relevant resources for all antigens and antibodies mentioned. This review provides a comprehensive summary of the different monoclonal antibodies that are currently in clinical use primarily in malignancy, including their function, which is of importance to those in the medical field and allied specialties.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Immunologic Factors/therapeutic use , Recombinant Proteins/therapeutic use
20.
Heart Fail Clin ; 18(3): 443-454, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35718418

ABSTRACT

Chimeric antigen receptor (CAR) T-cell and bispecific T-cell engager (BiTE) therapies have revolutionized the treatment of refractory or relapsed leukemia and lymphoma. Increased use of these therapies has revealed signals of significant cardiotoxicity, including cardiomyopathy/heart failure, arrhythmia, myocardial injury, hemodynamic instability, and cardiovascular death mainly in the context of a profound inflammatory response to CAR T-cell antitumor effects known as cytokine release syndrome (CRS). Preexisting cardiovascular risk factors and disease may increase the risk of such cardiotoxicity. High index of suspicion and close monitoring is required for prompt recognition. Supportive hemodynamic care and targeted anti-IL-6 therapy, as well as possibly broader immunosuppression with corticosteroids, are the cornerstones of the management.


Subject(s)
Cardiovascular Diseases , Receptors, Chimeric Antigen , Cardiotoxicity , Cardiovascular Diseases/therapy , Humans , Immunotherapy , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes
SELECTION OF CITATIONS
SEARCH DETAIL