Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.297
Filter
2.
Drug Metab Dispos ; 50(1): 8-16, 2022 01.
Article in English | MEDLINE | ID: mdl-34649966

ABSTRACT

Intratumoral heterogeneity is a leading cause of treatment failure resulting in tumor recurrence. For the antibody-drug conjugate (ADC) ado-trastuzumab emtansine (T-DM1), two major types of resistance include changes in human epidermal growth factor receptor 2 (HER2) expression and reduced payload sensitivity, which is often exacerbated by heterogenous HER2 expression and ADC distribution during treatment. ADCs with bystander payloads, such as trastuzumab-monomethyl auristatin E (T-MMAE), can reach and kill adjacent cells with lower receptor expression that cannot be targeted directly with the ADC. Additionally, coadministration of T-DM1 with its unconjugated antibody, trastuzumab, can improve distribution and minimize heterogeneous delivery. However, the effectiveness of trastuzumab coadministration and ADC bystander killing in heterogenous tumors in reducing the selection of resistant cells is not well understood. Here, we use an agent-based model to predict outcomes with these different regimens. The simulations demonstrate that both T-DM1 and T-MMAE benefit from trastuzumab coadministration for tumors with high average receptor expression (up to 70% and 40% decrease in average tumor volume, respectively), with greater benefit for nonbystander payloads. However, the benefit decreases as receptor expression is reduced, reversing at low concentrations (up to 360% and 430% increase in average tumor volume for T-DM1 and T-MMAE, respectively) for this mechanism that impacts both ADC distribution and efficacy. For tumors with intrinsic payload resistance, coadministration uniformly exhibits better efficacy than ADC monotherapy (50%-70% and 19%-36% decrease in average tumor volume for T-DM1 and T-MMAE, respectively). Finally, we demonstrate that several regimens select for resistant cells at clinical tolerable doses, which highlights the need to pursue other mechanisms of action for durable treatment responses. SIGNIFICANCE STATEMENT: Experimental evidence demonstrates heterogeneity in the distribution of both the antibody-drug conjugate and the target receptor in the tumor microenvironment, which can promote the selection of resistant cells and lead to recurrence. This study quantifies the impact of increasing the antibody dose and utilizing bystander payloads in heterogeneous tumors. Alternative cell-killing mechanisms are needed to avoid enriching resistant cell populations.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Immunotoxins/therapeutic use , Receptor, ErbB-2/genetics , Ado-Trastuzumab Emtansine , Aminobenzoates/therapeutic use , Cell Line, Tumor , Female , Humans , Immunoconjugates , Immunotherapy , Immunotoxins/pharmacokinetics , Models, Biological , Oligopeptides/therapeutic use , Trastuzumab/therapeutic use , Treatment Outcome , Xenograft Model Antitumor Assays
3.
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
4.
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
8.
Cancer Med ; 9(13): 4756-4767, 2020 07.
Article in English | MEDLINE | ID: mdl-32368864

ABSTRACT

INTRODUCTION: Ovarian cancer has only a 17% 5-year survival rate in patients diagnosed with late stage disease. Tumor-associated glycoprotein-72 (TAG72), expressed in 88% of all stages of ovarian cancer, is an excellent candidate for antibody-targeted therapy, as it is not expressed in normal human adult tissues, except in the secretory endometrium. METHODS: Using the clinically relevant anti-TAG72 murine monoclonal antibody CC49, we evaluated antibody drug conjugates (ADCs) incorporating the highly potent, synthetic antimitotic agent monomethylauristatin E (MMAE). MMAE was conjugated to CC49 via reduced disulfides in the hinge region, using three different types of linker chemistry, vinylsulfone (VS-MMAE), bromoacetamido (Br-MMAE), and maleimido (mal-MMAE). RESULTS: The drug antibody ratios (DARs) of the three ADCs were 2.3 for VS-MMAE, 10 for Br-MMAE, and 9.5 for mal-MMAE. All three ADCs exhibited excellent tumor to blood ratios on PET imaging, but the absolute uptake of CC49-mal-MMAE (3.3%ID/g) was low compared to CC49-Br-MMAE (6.43%ID/g), at 142 hours. Blood clearance at 43 hours was 38% for intact CC49, about 24% for both CC49-VS-MMAE and CC49-Br-MMAE, and 7% for CC49-mal-MMAE. CC49-VS-MMAE was not further studied due to its low DAR, while CC49-mal-MMAE was ineffective in the OVCAR3 xenograft likely due to its rapid blood clearance. In contrast, CC49-Br-MMAE treated mice exhibited an average of a 15.6 day tumor growth delay and a 40% increase in survival vs controls with four doses of 7.5 or 15 mg/kg of CC49-Br-MMAE. CONCLUSION: We conclude that CC49-Br-MMAE with a high DAR and stable linker performs well in a difficult to treat solid tumor model.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Antigens, Neoplasm/immunology , Immunoconjugates/therapeutic use , Immunologic Factors/therapeutic use , Oligopeptides/therapeutic use , Ovarian Neoplasms/therapy , Acetates/chemistry , Animals , Antibodies, Neoplasm/blood , Antibodies, Neoplasm/metabolism , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Female , HT29 Cells , Humans , Immunologic Factors/pharmacokinetics , Mice , Oligopeptides/blood , Oligopeptides/chemistry , Oligopeptides/metabolism , Ovarian Neoplasms/blood , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/metabolism , Positron-Emission Tomography , Random Allocation , Sulfones/chemistry
9.
Expert Rev Hematol ; 13(4): 331-349, 2020 04.
Article in English | MEDLINE | ID: mdl-32153224

ABSTRACT

Introduction: In the last few years, monoclonal antibodies have rapidly modified the therapeutic strategies for treating patients with multiple myeloma.Areas covered: In this review, the most recent literature data regarding indications for which monoclonal antibodies are currently or will be shortly approved as salvage therapies in relapsed/refractory myeloma are discussed. In particular, updated results until March 22, 2020 of antibodies directed against CD38 (daratumumab and isatuximab), SLAMF7 (elotuzumab), BCMA (GSK2857916/belantamab mafodotin), and PD-1/PD-1 L axis (nivolumab and pembrolizumab) will be analyzed in detail.Expert opinion: Monoclonal antibodies represent a new, very effective approach that will open novel and dynamic treatment scenarios for myeloma patients in the coming years. Optimal positioning and selection of different antibodies that are or will be soon available, appropriate combinations and careful evaluation of possible new toxicities should be considered in the future management of these patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Multiple Myeloma/drug therapy , Antibodies, Monoclonal/adverse effects , Antibodies, Neoplasm/adverse effects , Clinical Trials as Topic , Female , Humans , Male
10.
J Drug Target ; 28(6): 574-584, 2020 07.
Article in English | MEDLINE | ID: mdl-32037905

ABSTRACT

Cancer has become one of the major threats to human survival. Because of antibodies specificity and low toxicity, it is the primary choice to diagnose and treat cancer. It is easy to be cleared from the blood circulation or distributing throughout the body and causes unnecessary side effects. It is necessary to delivery antibodies to the tumour region in a stable, safe and effective manner. In this review, we discuss the latest studies that aimed to delivery antibodies to tumour sites via several vector forms, such as liposomes, carbon nanomaterials, and gold nanomaterials. How to deliver antibodies to the target site is a difficulty for antibody therapy. This review summarises the antibody's therapeutic forms and carrier materials in recent years, and to explore how antibodies can be safely and stably delivered to the target site.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Drug Delivery Systems , Neoplasms/therapy , Animals , Drug Carriers , Humans
11.
Front Immunol ; 11: 622442, 2020.
Article in English | MEDLINE | ID: mdl-33569063

ABSTRACT

The contributions of the humoral immune response to melanoma are now widely recognized, with reports of positive prognostic value ascribed to tumor-infiltrating B cells (TIL-B) and increasing evidence of B cells as key predictors of patient response to treatment. There are disparate views as to the pro- and anti-tumor roles of B cells. B cells appear to play an integral role in forming tumor-associated tertiary lymphoid structures (TLSs) which can further modulate T cell activation. Expressed antibodies may distinctly influence tumor regulation in the tumor microenvironment, with some isotypes associated with strong anti-tumor immune response and others with progressive disease. Recently, B cells have been evaluated in the context of cancer immunotherapy. Checkpoint inhibitors (CPIs), targeting T cell effector functions, have revolutionized the management of melanoma for many patients; however, there remains a need to accurately predict treatment responders. Increasing evidence suggests that B cells may not be simple bystanders to CPI immunotherapy. Mature and differentiated B cell phenotypes are key positive correlates of CPI response. Recent evidence also points to an enrichment in activatory B cell phenotypes, and the contribution of B cells to TLS formation may facilitate induction of T cell phenotypes required for response to CPI. Contrastingly, specific B cell subsets often correlate with immune-related adverse events (irAEs) in CPI. With increased appreciation of the multifaceted role of B cell immunity, novel therapeutic strategies and biomarkers can be explored and translated into the clinic to optimize CPI immunotherapy in melanoma.


Subject(s)
Antibodies, Neoplasm/therapeutic use , B-Lymphocytes , Immune Checkpoint Inhibitors/therapeutic use , Melanoma , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Humans , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy
12.
JCI Insight ; 5(2)2020 01 30.
Article in English | MEDLINE | ID: mdl-31855578

ABSTRACT

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


Subject(s)
Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , B-Lymphocytes/immunology , Lymph Nodes/drug effects , Neoplasms/immunology , Programmed Cell Death 1 Receptor/drug effects , T-Lymphocytes/immunology , Animals , Antibodies, Neoplasm/therapeutic use , Female , Interferon-gamma/metabolism , Interleukin-2/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Natural Killer T-Cells , Neoplasms/drug therapy , Proteomics , Tumor Necrosis Factor-alpha/metabolism
13.
Clin Transl Oncol ; 22(7): 1094-1104, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31732916

ABSTRACT

BACKGROUND: T cell therapy for cancer involves genetic introduction of a target-binding feature into autologous T cells, ex vivo expansion and single large bolus administration back to the patient. These reprogrammed T cells can be highly effective in killing cells, but tumor heterogeneity results in regrowth of cells that do not sufficiently express the single antigen being targeted. We describe a cell-based therapy that simultaneously targets multiple tumor-specific antigens. METHODS: High-affinity polyclonal rabbit antibodies were generated against nine different surface-related tumor-specific mutations on B16F10 cells. Unsorted splenic effector cells from syngeneic mice were incubated with a cocktail of the nine anti-B16F10 antibodies. These 'armed' effector cells were used to treat mice previously inoculated with B16F10 melanoma cells. RESULTS: The cocktail of nine antibodies resulted in dense homogeneous binding to histological sections of B16F10 cells. Five treatments with the armed effector cells and PD1 inhibition inhibited tumor growth and improved survival. Shortening the interval of the five treatments from every three days to every day increased survival. Arming effector cells with the four antibodies showing best binding to B16F10 cells even further increased survival. CONCLUSIONS: This study demonstrates that ex vivo arming a mixed population of immune effector cells with antibodies targeting multiple tumor-specific mutated proteins in conjunction with PD1 inhibition delayed tumor growth and prolonged survival in mice inoculated with an aggressive melanoma. A remarkably low total antibody dose of less than 5 µg was sufficient to accomplish tumor inhibition. Scaling up to clinical level may be feasible.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/methods , Leukocytes/immunology , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Tumor Burden , Animals , Antigens, Neoplasm/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mutant Proteins/genetics , Mutant Proteins/immunology , Mutation , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Spleen/cytology , Survival Rate
14.
J Autoimmun ; 104: 102310, 2019 11.
Article in English | MEDLINE | ID: mdl-31421963

ABSTRACT

Advances in our understanding οf tumor immunity have prompted a paradigm shift in oncology, with the emergence of immunotherapy, where therapeutic agents are used to target immune cells rather than cancer cells. A real breakthrough in the field of immunotherapy came with the use of immune checkpoint inhibitors (ICI), namely antagonistic antibodies that block key immune regulatory molecules (checkpoint molecules), such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death protein (PD-1) and its ligand PD-L1, that under physiologic conditions suppress T cell effector function. However, despite the enormous success, a significant proportion of patients do not respond, while responses are frequently accompanied by life-threatening autoimmune related adverse events (irAEs). A major impediment in the effectiveness of ICI immunotherapy is the tumoral resistance, which is dependent on the immunosuppressive nature of tumor microenvironment (TME). Regulatory T cells (Tregs) are among the most abundant suppressive cells in the TME and their presence has been correlated with tumor progression, invasiveness as well as metastasis. Tregs are characterized by the expression of the transcription factor Foxp3 and various mechanisms ranging from cell-to-cell contact to secretion of inhibitory molecules have been implicated in their function. Notably, Tregs amply express most of the checkpoint molecules such as CTLA4, PD1 and LAG3 and therefore represent a direct target of ICI immunotherapy. Taking into consideration the critical role of Tregs in maintenance of immune homeostasis and avoidance of autoimmunity it is plausible that targeting of Tregs by ICI immunotherapy results in the development of irAEs. Since the use of ICI becomes common, and new immune checkpoint molecules are currently under clinical trials for the treatment of cancer, the occurrence of irAEs is expected to dramatically rise. Herein we review the current literature focusing on the role of Tregs in cancer evolution, ICI response and development of irAEs. Unraveling the complex mechanisms that hinder the tumor immune surveillance and in particular how ICI immunotherapy imprint on Treg activities to promote cancer regression while avoid development of irAEs, will empower the design of novel immunotherapeutic modalities in cancer with increased efficacy and diminished adverse events.


Subject(s)
Antibodies, Neoplasm , Autoimmune Diseases , Immunotherapy/adverse effects , Neoplasms , T-Lymphocytes, Regulatory/immunology , Antibodies, Neoplasm/adverse effects , Antibodies, Neoplasm/therapeutic use , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/therapy , Humans , Neoplasm Proteins/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes, Regulatory/pathology
15.
Database (Oxford) ; 20192019 01 01.
Article in English | MEDLINE | ID: mdl-31169290

ABSTRACT

The clinical efficacy of therapeutic monoclonal antibodies for breast and colorectal cancer has greatly contributed to the improvement of patients' outcomes by individualizing their treatments according to their genomic background. However, primary or acquired resistance to treatment reduces its efficacy. In this context, the identification of biomarkers predictive of drug response would support research and development of new alternative treatments. Biomarkers play a major role in the genomic revolution, supporting disease diagnosis and treatment decision-making. Currently, several molecular biomarkers of treatment response for breast and colorectal cancer have been described. However, information on these biomarkers is scattered across several resources, and needs to be identified, collected and properly integrated to be fully exploited to inform monitoring of drug response in patients. Therefore, there is a need of resources that offer biomarker data in a harmonized manner to the user to support the identification of actionable biomarkers of response to treatment in cancer. ResMarkerDB was developed as a comprehensive resource of biomarkers of drug response in colorectal and breast cancer. It integrates data of biomarkers of drug response from existing repositories, and new data extracted and curated from the literature (referred as ResCur). ResMarkerDB currently features 266 biomarkers of diverse nature. Twenty-five percent of these biomarkers are exclusive of ResMarkerDB. Furthermore, ResMarkerDB is one of the few resources offering non-coding DNA data in response to drug treatment. The database contains more than 500 biomarker-drug-tumour associations, covering more than 100 genes. ResMarkerDB provides a web interface to facilitate the exploration of the current knowledge of biomarkers of response in breast and colorectal cancer. It aims to enhance translational research efforts in identifying actionable biomarkers of drug response in cancer.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Biomarkers, Tumor , Breast Neoplasms , Colorectal Neoplasms , Databases, Factual , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Male
16.
Front Immunol ; 10: 481, 2019.
Article in English | MEDLINE | ID: mdl-30930905

ABSTRACT

Immunotherapy with therapeutic antibodies has increased survival for patients with hematologic and solid cancers. Still, a significant fraction of patients fails to respond to therapy or acquire resistance. Understanding and overcoming mechanisms of resistance to antibody drugs, and in particular those common to antibody drugs as a class, is therefore highly warranted and holds promise to improve response rates, duration of response and potentially overall survival. Activating and inhibitory Fc gamma receptors (FcγR) are known to coordinately regulate therapeutic activity of tumor direct-targeting antibodies. Similar, but also divergent, roles for FcγRs in controlling efficacy of immune modulatory antibodies e.g., checkpoint inhibitors have been indicated from mouse studies, and were recently implicated in contributing to efficacy in the human clinical setting. Here we discuss evidence and mechanisms by which Fc gamma receptors-the "antibody checkpoints"-regulate antibody-induced antitumor immunity. We further discuss how targeted blockade of the sole known inhibitory antibody checkpoint FcγRIIB may help overcome resistance and boost activity of clinically validated and emerging antibodies in cancer immunotherapy.


Subject(s)
Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/immunology , Drug Resistance, Neoplasm , Humans , Mice , Neoplasms/immunology , Tumor Microenvironment/immunology
17.
Immunol Lett ; 205: 31-39, 2019 01.
Article in English | MEDLINE | ID: mdl-29758241

ABSTRACT

The ectonucleotidases CD39 and CD73 are cell surface enzymes that catabolize the breakdown of extracellular ATP into adenosine. As such, they constitute critical components of the extracellular purinergic pathway and play important roles in maintaining tissue and immune homeostasis. With the coming of age of cancer immunotherapy, ectonucleotidases and adenosine receptors have emerged as novel therapeutic targets to enhance antitumor immune responses. With early-phase clinical trials showing promising results, it is becoming increasingly important to decipher the distinct mechanisms-of-action of adenosine-targeting agents, identify patients that will benefit from these agents and rationally develop novel synergistic combinations. Given the broad expression of ectonucleotidases and adenosine receptors, a better understanding of cell-specific roles will also be key for successful implementation of this new generation of immuno-oncology therapeutics. We here review the latest studies on the roles of CD73 and adenosine in cancer with a focus on cell-specific function. We also discuss ongoing clinical trials and future avenues for adenosine-targeting agents.


Subject(s)
5'-Nucleotidase/immunology , Adenosine/immunology , Antibodies, Neoplasm/immunology , Neoplasms/immunology , Purinergic Antagonists/immunology , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Animals , Antibodies, Neoplasm/therapeutic use , Gene Expression Regulation, Neoplastic/immunology , Humans , Neoplasms/therapy , Purinergic Antagonists/therapeutic use , Receptors, Purinergic P1/immunology , Receptors, Purinergic P1/metabolism , Signal Transduction/immunology
18.
Curr Top Med Chem ; 18(13): 1091-1109, 2018.
Article in English | MEDLINE | ID: mdl-30068276

ABSTRACT

Cancer has become one of the main leading causes of morbidity and mortality worldwide. One of the critical drawbacks of current cancer therapeutics has been the lack of the target-selectivity, as these drugs should have an effect exclusively on cancer cells while not perturbing healthy ones. In addition, their mechanism of action should be sufficiently fast to avoid the invasion of neighbouring healthy tissues by cancer cells. The use of conventional chemotherapeutic agents and other traditional therapies, such as surgery and radiotherapy, leads to off-target interactions with serious side effects. In this respect, recently developed target-selective Antibody-Drug Conjugates (ADCs) are more effective than traditional therapies, presumably due to their modular structures that combine many chemical properties simultaneously. In particular, ADCs are made up of three different units: a highly selective Monoclonal antibody (Mab) which is developed against a tumour-associated antigen, the payload (cytotoxic agent), and the linker. The latter should be stable in circulation while allowing the release of the cytotoxic agent in target cells. The modular nature of these drugs provides a platform to manipulate and improve selectivity and the toxicity of these molecules independently from each other. This in turn leads to generation of second- and third-generation ADCs, which have been more effective than the previous ones in terms of either selectivity or toxicity or both. Development of ADCs with improved efficacy requires knowledge at the atomic level regarding the structure and dynamics of the molecule. As such, we reviewed all the most recent computational methods used to attain all-atom description of the structure, energetics and dynamics of these systems. In particular, this includes homology modelling, molecular docking and refinement, atomistic and coarse-grained molecular dynamics simulations, principal component and cross-correlation analysis. The full characterization of the structure-activity relationship devoted to ADCs is critical for antibody-drug conjugate research and development.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Design , Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Humans , Molecular Docking Simulation
19.
Mol Biol (Mosk) ; 52(3): 380-393, 2018.
Article in Russian | MEDLINE | ID: mdl-29989572

ABSTRACT

Bispecific antibodies capable of simultaneously binding two targets have been studied for many years with a view to their implementation in clinical practice. Unique biological and pharmacological properties, as well as the diversity of their formats, make it possible to consider bispecific antibodies as promising agents for use in various procedures: from visualization of intracellular processes to targeted anticancer therapy. Bispecific antibodies help to determine more precisely the therapeutic target, thereby increasing the efficiency of therapy and reducing the probability of side effects. The present review describes the main formats of bispecific antibodies, methods for their generation, and possibilities for practical application.


Subject(s)
Antibodies, Bispecific , Antibodies, Neoplasm , Neoplasms , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy
20.
Cell Physiol Biochem ; 47(2): 441-457, 2018.
Article in English | MEDLINE | ID: mdl-29794415

ABSTRACT

The accumulation of mutated somatic cells due to the incompetency of body's immune system may lead to tumor onset. Therefore, enhancing the ability of the system to eliminate such cells should be the core of tumor therapy. The intrinsic antitumor immunity is triggered by tumor-specific antigens (TSA) or TSA-sensitized dendritic cells (DC). Once initiated, specific anti-tumor antibodies are produced and tumor-specific killer immune cells, including cytotoxic T lymphocytes (CTL), NK cells, and macrophages, are raised or induced. Several strategies may enhance antitumor action of immune system, such as supplying tumor-targeted antibody, activating T cells, enhancing the activity and tumor recognition of NK cells, promoting tumor-targeted phagocytosis of macrophages, and eliminating the immunosuppressive myeloid-derived suppressor cells (MDSCs) and Treg cells. Apart from the immune system, the removal of tumor burden still needs to be assisted by drugs, surgery or radiation. And the body's internal environment and tumor microenvironment should be improved to recover immune cell function and prevent tumor growth. Multiple microenvironment modulatory therapies may be applied, including addressing hypoxia and oxidative stress, correcting metabolic disorders, and controlling chronic inflammation. Finally, to cure tumor and prevent tumor recurrence, repairing or supporting therapy that consist of tissue repair and nutritional supplement should be applied properly.


Subject(s)
Immunotherapy , Neoplasms/therapy , Antibodies, Neoplasm/therapeutic use , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immune System/cytology , Immune System/metabolism , Killer Cells, Natural/immunology , Neoplasms/metabolism , Neoplasms/pathology , Oxidative Stress , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...