Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Biomed Eng ; 8(2): 193-200, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37996615

ABSTRACT

Owing to the immunogenicity of adeno-associated viruses (AAVs), gene therapies using AAVs face considerable obstacles. Here, by leveraging ex vivo T-cell assays, the prediction of epitope binding to major histocompatibility complex class-II alleles, sequence-conservation analysis in AAV phylogeny and site-directed mutagenesis, we show that the replacement of amino acid residues in a promiscuous and most immunodominant T-cell epitope in the AAV9 capsid with AAV5 sequences abrogates the immune responses of peripheral blood mononuclear cells to the chimaeric vector while preserving its functions, potency, cellular specificity, transduction efficacy and biodistribution. This rational approach to the immunosilencing of capsid epitopes promiscuously binding to T cells may be applied to other AAV vectors and epitope regions.


Subject(s)
Capsid , Dependovirus , Capsid/chemistry , Capsid/metabolism , Dependovirus/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/analysis , Epitopes, T-Lymphocyte/metabolism , Leukocytes, Mononuclear , Tissue Distribution , Capsid Proteins/genetics , Capsid Proteins/chemistry , Capsid Proteins/metabolism
2.
Front Immunol ; 14: 1211529, 2023.
Article in English | MEDLINE | ID: mdl-37469509

ABSTRACT

During gene therapy trials, immune responses against adeno-associated virus (AAV) vectors are monitored by antibody assays that detect the humoral and T-cell mediated cellular responses to AAV vectors. T cell assays commonly utilize the collection of patients' peripheral blood mononuclear cells (PBMCs) and stimulation with AAV-derived overlapping peptides. We recently described that spontaneous deamidation coincides with T cell epitopes in AAV capsids and that spontaneous deamidation may enhance or decrease immunogenicity in some individuals. This raised the concern for false negative results of antibody detection and PBMC immune monitoring assays because these assays use wild-type (WT) AAV or WT peptides for T cell re-stimulation and these peptides may not re-activate T cells that were stimulated with deamidated AAV capsid. To investigate this concern, we modeled the scenario by expanding T cells with deamidated peptides and evaluated the cross-reactivity of expanded T cells to WT peptides. In the majority of samples, cells that were expanded with deamidated peptides and restimulated with WT peptide had significantly lowered IL-2 and IFN-γ production. Spiking the four deamidated peptides to the WT peptide pool used for re-stimulation, restored the signal and corrected the performance of the assay. We also evaluated the impact of deamidation on anti AAV binding antibodies and did not observe a major impact on seroprevalence detection of AAV9. These data indicate that a high level of deamidation in AAV therapy may result in underestimation or even failure to detect immune responses against WT peptides during cellular immune monitoring.


Subject(s)
Leukocytes, Mononuclear , T-Lymphocytes , Humans , Monitoring, Immunologic , Seroepidemiologic Studies , Dependovirus , Peptides/metabolism
3.
Mol Ther Methods Clin Dev ; 24: 255-267, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35211638

ABSTRACT

Despite the high safety profile demonstrated in clinical trials, the immunogenicity of adeno-associated virus (AAV)-mediated gene therapy remains a major hurdle. Specifically, T-cell-mediated immune responses to AAV vectors are related to loss of efficacy and potential liver toxicities. As post-translational modifications in T cell epitopes have the potential to affect immune reactions, the cellular immune responses to peptides derived from spontaneously deamidated AAV were investigated. Here, we report that highly deamidated sites in AAV9 contain CD4 T cell epitopes with a Th1 cytokine pattern in multiple human donors with diverse human leukocyte antigen (HLA) backgrounds. Furthermore, some peripheral blood mononuclear cell (PBMC) samples demonstrated differential T cell activation to deamidated or non-deamidated epitopes. Also, in vitro and in silico HLA binding assays showed differential binding to the deamidated or non-deamidated peptides in some HLA alleles. This study provides critical attributes to vector-immune-mediated responses, as AAV deamidation can impact the immunogenicity, safety, and efficacy of AAV-mediated gene therapy in some patients.

4.
Commun Biol ; 4(1): 1048, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34497355

ABSTRACT

In a biologic therapeutic landscape that requires versatility in targeting specificity, valency and half-life modulation, the monomeric Fc fusion platform holds exciting potential for the creation of a class of monovalent protein therapeutics that includes fusion proteins and bispecific targeting molecules. Here we report a structure-guided approach to engineer monomeric Fc molecules to adapt multiple versions of half-life extension modifications. Co-crystal structures of these monomeric Fc variants with Fc neonatal receptor (FcRn) shed light into the binding interactions that could serve as a guide for engineering the half-life of antibody Fc fragments. These engineered monomeric Fc molecules also enabled the generation of a novel monovalent bispecific molecular design, which translated the FcRn binding enhancement to improvement of in vivo serum half-life.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunoglobulin Fc Fragments/metabolism , Receptors, Fc/metabolism , Animals , Half-Life , Histocompatibility Antigens Class I/pharmacology , Humans , Immunoglobulin Fc Fragments/pharmacology , Mice , Mice, Transgenic , Protein Engineering
5.
Front Immunol ; 11: 1261, 2020.
Article in English | MEDLINE | ID: mdl-32695104

ABSTRACT

Immunotoxins are cytolytic fusion proteins developed for cancer therapy, composed of an antibody fragment that binds to a cancer cell and a protein toxin fragment that kills the cell. Pseudomonas exotoxin A (PE) is a potent toxin that is used for the killing moiety in many immunotoxins. Moxetumomab Pasudotox (Lumoxiti) contains an anti-CD22 Fv and a 38 kDa portion of PE. Lumoxiti was discovered in the Laboratory of Molecular Biology at the U.S. National Cancer Institute and co-developed with Medimmune/AstraZeneca to treat hairy cell leukemia. In 2018 Lumoxiti was approved by the US Food and Drug Administration for the treatment of drug-resistant Hairy Cell Leukemia. Due to the bacterial origin of the killing moiety, immunotoxins containing PE are highly immunogenic in patients with normal immune systems, but less immunogenic in patients with hematologic malignancies, whose immune systems are often compromised. LMB-100 is a de-immunized variant of the toxin with a humanized antibody that targets mesothelin and a PE toxin that was rationally designed for diminished reactivity with antibodies and B cell receptors. It is now being evaluated in clinical trials for the treatment of mesothelioma and pancreatic cancer and is showing somewhat diminished immunogenicity compared to its un modified parental counterpart. Here we review the immunogenicity of the original and de-immunized PE immunotoxins in mice and patients, the development of anti-drug antibodies (ADAs), their impact on drug availability and their effect on clinical efficacy. Efforts to mitigate the immunogenicity of immunotoxins and its impact on immunogenicity will be described including rational design to identify, remove, or suppress B cell or T cell epitopes, and combination of immunotoxins with immune modulating drugs.


Subject(s)
Exotoxins/immunology , Immunotoxins/immunology , Pseudomonas/immunology , ADP Ribose Transferases/immunology , Animals , Antibody Formation/immunology , Bacterial Toxins/immunology , Clinical Trials as Topic , Epitopes, B-Lymphocyte/immunology , Exotoxins/chemistry , Exotoxins/pharmacokinetics , Humans , Immunoassay , Immunomodulation/drug effects , Immunotoxins/chemistry , Immunotoxins/pharmacokinetics , Mesothelin , Neoplasms/drug therapy , Recombinant Proteins/immunology , Structure-Activity Relationship , Treatment Outcome , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
6.
Toxins (Basel) ; 10(11)2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30441807

ABSTRACT

SS1P is an anti-mesothelin immunotoxin composed of a targeting antibody fragment genetically fused to a truncated fragment of Pseudomonas exotoxin A. Delayed responses reported in mesothelioma patients receiving SS1P suggest that anti-tumor immunity is induced. The goal of this study is to evaluate if SS1P therapy renders mesothelioma tumors more sensitive to cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint blockade. We evaluated the ability of SS1P to induce adenosine triphosphate (ATP) secretion and calreticulin expression on the surface of AE17M mouse mesothelioma cells. Both properties are associated with immunogenic cell death. Furthermore, we treated these tumors with intra-tumoral SS1P and systemic CTLA-4. We found that SS1P increased the release of ATP from AE17M cells in a dose and time-dependent manner. In addition, SS1P induced calreticulin expression on the surface of AE17M cells. These results suggest that SS1P promotes immunogenic cell death and could sensitize tumors to anti-CTLA-4 based therapy. In mouse studies, we found that the combination of anti-CTLA-4 with intra-tumoral SS1P induced complete regressions in most mice and provided a statistically significant survival benefit compared to monotherapy. The surviving mice were protected from tumor re-challenge, indicating the development of anti-tumor immunity. These findings support the use of intra-tumoral SS1P in combination with anti-CTLA-4.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , CTLA-4 Antigen/antagonists & inhibitors , Mesothelioma/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Female , Mesothelin , Mesothelioma/pathology , Mice, Inbred C57BL , Tumor Burden/drug effects
7.
Cell Immunol ; 334: 38-41, 2018 12.
Article in English | MEDLINE | ID: mdl-30213644

ABSTRACT

LMB-100 is a recombinant immunotoxin being developed for cancer treatment that is composed of a Fab that binds to mesothelin and a portion of Pseudomonas exotoxin A. LMB-100 is in clinical trials for the treatment of mesothelioma and pancreatic cancer. To determine if check point modulating antibodies enhance the formation of anti-drug antibodies (ADA) against LMB-100, we treated mice with LMB-100 and four different immune modulating monoclonal antibodies that have different mechanisms of action; anti-CTLA4, anti-OX40, anti-PD-1 and anti-PDL-1. We found that anti-PD-1 and anti PDL-1 do not increase the formation of ADA, but anti-CTLA-4 and anti-OX-40 do increase the onset of ADA. These results indicate that combining anti-CTLA-4 and anti-OX-40 with antibodies and other protein-based therapeutics may enhance ADA formation in humans.


Subject(s)
Antibodies, Monoclonal/immunology , B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Immunotoxins/immunology , Programmed Cell Death 1 Receptor/immunology , Receptors, OX40/immunology , Animals , Female , GPI-Linked Proteins/immunology , Mesothelin , Mice , Mice, Inbred BALB C
8.
Am J Pathol ; 188(8): 1736-1743, 2018 08.
Article in English | MEDLINE | ID: mdl-29870741

ABSTRACT

Recombinant immunotoxins (RITs) are genetically engineered proteins being developed to treat cancer. They are composed of an Fv that targets a cancer antigen and a fragment of a bacterial toxin that kills tumor cells. Because the toxin is a foreign protein, it is immunogenic. The clinical success of RITs in patients with a normal immune system is limited by their immunogenicity. In this review, we discuss our progress in therapeutic protein deimmunization and the balancing act between immunogenicity and therapeutic potency. One approach is to prevent the activation of B cells by mapping and elimination of B-cell epitopes. A second approach is to prevent helper T-cell activation by interfering with major histocompatibility complex II presentation or T-cell recognition. Immunizing mice with RITs that were deimmunized by elimination of the murine B- or T-cell epitopes showed that both approaches are effective. Another approach to control immunogenicity is to modify the host immune system. Nanoparticles containing synthetic vaccine particles encapsulating rapamycin can induce immune tolerance and prevent anti-drug antibody formation. This treatment restores RIT anti-tumor activity that is otherwise neutralized because of immunogenicity.


Subject(s)
Immunotherapy , Immunotoxins/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Recombinant Proteins/therapeutic use , Animals , Humans
9.
Mol Cancer Ther ; 17(7): 1486-1493, 2018 07.
Article in English | MEDLINE | ID: mdl-29695631

ABSTRACT

Tac (CD25) is expressed on multiple hematologic malignancies and is a target for cancer therapies. LMB-2 is an extremely active anti-Tac recombinant immunotoxin composed of an Fv that binds to Tac and a 38-kDa fragment of Pseudomonas exotoxin A (PE38). Although LMB-2 has shown high cytotoxicity toward Tac-expressing cancer cells in clinical trials, its efficacy was hampered by the formation of anti-drug antibodies against the immunogenic bacterial toxin and by dose-limiting off-target toxicity. To reduce toxin immunogenicity and nonspecific toxicity, we introduced six point mutations into domain III that were previously shown to reduce T-cell immunogenicity and deleted domain II from the toxin, leaving only the 11aa furin cleavage site, which is required for cytotoxic activity. Although this strategy has been successfully implemented for mesothelin and CD22-targeting immunotoxins, we found that removal of domain II significantly lowered the cytotoxic activity of anti-Tac immunotoxins. To restore cytotoxic activity in the absence of PE domain II, we implemented a combined rational design and screening approach to isolate highly active domain II-deleted toxin variants. The domain II-deleted variant with the highest activity contained an engineered disulfide-bridged furin cleavage site designed to mimic its native conformation within domain II. We found that this approach restored 5-fold of the cytotoxic activity and dramatically improved the MTD. Both of these improvements led to significantly increased antitumor efficacy in vivo We conclude that the next-generation anti-Tac immunotoxin is an improved candidate for targeting Tac-expressing malignancies. Mol Cancer Ther; 17(7); 1486-93. ©2018 AACR.


Subject(s)
ADP Ribose Transferases/administration & dosage , Antibodies, Monoclonal/administration & dosage , Bacterial Toxins/administration & dosage , Exotoxins/administration & dosage , Hematologic Neoplasms/drug therapy , Protein Engineering , Virulence Factors/administration & dosage , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/genetics , Exotoxins/genetics , Exotoxins/immunology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Humans , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-2 Receptor alpha Subunit/immunology , Mesothelin , Mice , Point Mutation/genetics , Protein Domains/genetics , Protein Domains/immunology , Sialic Acid Binding Ig-like Lectin 2/antagonists & inhibitors , Sialic Acid Binding Ig-like Lectin 2/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Virulence Factors/genetics , Virulence Factors/immunology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
10.
J Immunol ; 200(6): 2038-2045, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29431691

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins being developed for cancer treatment. They are composed of an Ab fragment that targets a cancer Ag and a cytotoxic portion of Pseudomonas exotoxin A. They are effective for patients with hematologic malignancies with defective immunity, but their efficacy against solid tumors is limited by anti-drug Ab (ADA) responses in immune-competent patients. Pre-existing Abs or immune memory owing to previous toxin exposure represent additional hurdles because they induce rapid and strong ADA responses. Here, we evaluated the efficacy of methotrexate (MTX) to prevent ADA formation against the mesothelin-targeting RIT LMB-100 in naive mice and in mice with pre-existing Abs. We found that low-dose MTX combined with LMB-100 completely suppressed the formation of ADAs in a dose- and frequency-dependent manner. Suppression of the immune response restored blood levels of LMB-100 and prevented its neutralization. Furthermore, combination of MTX with LMB-100 did not compromise the immune response against a second Ag given after stopping MTX, indicating specific immune tolerance. Adoptive transfer of splenocytes suppressed Ab responses to LMB-100 in recipient mice, indicating a durable immune tolerance. We conclude that combination of MTX and LMB-100 is effective at preventing immune responses in a durable, Ag-specific manner. We propose combining low-dose MTX in immune-competent cancer patients receiving RIT therapy to prevent immunogenicity. This approach could be applied to other immunogenic therapeutic agents and to proteins for which there is pre-existing immunity.


Subject(s)
Immune Tolerance/drug effects , Immunity, Humoral/drug effects , Immunotoxins/immunology , Methotrexate/pharmacology , Recombinant Proteins/immunology , ADP Ribose Transferases/immunology , Adoptive Transfer/methods , Animals , Antibodies, Monoclonal/immunology , Antibody Formation/drug effects , Bacterial Toxins/immunology , Cells, Cultured , Exotoxins/immunology , Female , GPI-Linked Proteins/pharmacology , Immune Tolerance/immunology , Immunity, Humoral/immunology , Immunotherapy/methods , Mesothelin , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
11.
Proc Natl Acad Sci U S A ; 115(4): E733-E742, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29311317

ABSTRACT

Protein-based drugs are very active in treating cancer, but their efficacy can be limited by the formation of neutralizing antidrug antibodies (ADAs). Recombinant immunotoxins are proteins that are very effective in patients with leukemia, where immunity is suppressed, but induce ADAs, which compromise their activity, in patients with intact immunity. Here we induced a specific, durable, and transferable immune tolerance to recombinant immunotoxins by combining them with nanoparticles containing rapamycin (SVP-R). SVP-R mitigated the formation of inhibitory ADAs in naïve and sensitized mice, resulting in restoration of antitumor activity. The immune tolerance is mediated by colocalization of the SVP-R and immunotoxin to dendritic cells and macrophages in the spleen and is abrogated by depletion of regulatory T cells. Tolerance induced by SVPs was not blocked by checkpoint inhibitors or costimulatory agonist monoclonal antibodies that by themselves enhance ADA formation.


Subject(s)
Immunomodulation , Immunosuppressive Agents/administration & dosage , Immunotoxins/administration & dosage , Leukemia/therapy , Sirolimus/administration & dosage , Animals , Antibodies, Neutralizing , GPI-Linked Proteins/immunology , Humans , Immunotoxins/immunology , Mesothelin , Nanoparticles , Time Factors
12.
Cell Immunol ; 313: 59-66, 2017 03.
Article in English | MEDLINE | ID: mdl-28087047

ABSTRACT

LMB-2, is a potent recombinant immunotoxin (RIT) that is composed of scFv antibody that targets CD25 (Tac) and a toxin fragment (PE38). It is used to treat T cell leukemias and lymphomas. To make LMB-2 less immunogenic, we introduced a large deletion in domain II and six point mutations in domain III that were previously shown to reduce T cell activation in other RITs. We found that unlike other RITs, deletion of domain II from LMB-2 severely compromised its activity. Rather than deletion, we identified T cell epitopes in domain II and used alanine substitutions to identify point mutations that diminished those epitopes. The novel RIT, LMB-142 contains a 38kDa toxin and nine point mutations that diminished T cell response to the corresponding peptides by an average of 75%. LMB-142 has good cytotoxic activity and has lower nonspecific toxicity in mice. LMB-142 should be more efficient in cancer therapy because more treatment cycles can be given.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Immunotoxins/therapeutic use , Leukemia, T-Cell/therapy , Pseudomonas/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/genetics , Bacterial Toxins/genetics , Cell Line, Tumor , Cell Proliferation , Cytokines/metabolism , Drug Design , Enzyme-Linked Immunospot Assay , Epitopes, T-Lymphocyte/genetics , Exotoxins/genetics , Exotoxins/therapeutic use , Female , Genetic Engineering , Humans , Immunotoxins/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Leukemia, T-Cell/immunology , Lymphocyte Activation , Mice , Mutagenesis, Site-Directed , Mutation/genetics
13.
Cell Mol Immunol ; 14(5): 432-442, 2017 May.
Article in English | MEDLINE | ID: mdl-26477977

ABSTRACT

Antibodies against the toxin portion of recombinant immunotoxins (RIT) reduce their efficacy and pose a potential safety risk. To overcome this problem we mutated the very immunogenic immunotoxin SS1P to produce LMB-T20, a de-immunized RIT that has the eight human T-cell epitopes in SS1P modified or removed. To determine the effect of T-cell epitope removal in vivo we mapped the T-cell epitopes in immune-competent BALB/c mice and found that these mice recognize two epitopes. One corresponds to the human immunodominant T-cell epitope and the other to a human subdominant epitope; both were eliminated in LMB-T20. We found that mice immunized with LMB-T20 did not have T-cell activation and did not develop anti-drug antibodies (ADA), whereas mice immunized with SS1P, showed T-cell activation, and developed ADA detected by both ELISA and drug neutralizing assays. The ability of the mice treated with LMB-T20 to respond to other antigens was not compromised. We conclude that elimination of T-cell epitopes is sufficient to prevent formation of antibodies to an immunogenic foreign protein.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies/immunology , Antibody Formation/immunology , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Female , Humans , Immunization , Lymphocyte Activation , Mice , Mice, Inbred BALB C
14.
AAPS J ; 19(1): 117-129, 2017 01.
Article in English | MEDLINE | ID: mdl-27796910

ABSTRACT

Identification of helper T-cell epitopes is important in many fields of medicine. We previously used an experimental approach to identify T-cell epitopes in PE38, a truncated bacterial toxin used in immunotoxins. Here, we evaluated the ability of antibodies to DR, DP, or DQ to block T-cell responses to PE38 epitopes in 36 PBMC samples. We predicted the binding affinities of peptides to DR, DP, and DQ alleles using computational tools and analyzed their ability to predict the T-cell epitopes. We found that HLA-DR is responsible for 65% of the responses, DP 24%, and DQ 4%. One epitope that is presented in 20% of the samples (10/50) is entirely DP restricted and was not predicted to bind to DR or DP reference alleles using binding algorithms. We conclude that DP has an important role in helper T-cell response to PE38.


Subject(s)
Bacterial Toxins/immunology , Epitopes, T-Lymphocyte/immunology , HLA-DP Antigens/chemistry , Immunotoxins/immunology , Peptides/chemistry , Algorithms , Alleles , Amino Acid Sequence , Binding Sites , Cells, Cultured , Epitopes, T-Lymphocyte/genetics , Flow Cytometry , Genes, MHC Class II , HLA-DP Antigens/genetics , HLA-DP Antigens/immunology , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Peptides/immunology
15.
Oncotarget ; 7(21): 29916-26, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27167198

ABSTRACT

Recombinant immunotoxins (RITs) are genetically engineered proteins being developed to treat cancer. They are composed of an Fv that targets a cancer antigen and a portion of a protein toxin. Their clinical success is limited by their immunogenicity. Our goal is to produce a new RIT that targets mesothelin and is non-immunogenic by combining mutations that decrease B- and T-cell epitopes. Starting with an immunotoxin that has B-cell epitopes suppressed, we added mutations step-wise that suppress T-cell epitopes. The final protein (LMB-T14) has greatly reduced antigenicity as assessed by binding to human anti-sera and a greatly decreased ability to activate helper T-cells evaluated in a T-cell activation assay. It is very cytotoxic to mesothelioma cells from patients, and to cancer cell lines. LMB-T14 produces complete remissions of a mesothelin expressing cancer (A431/H9) xenograft. The approach used here can be used to de-immunize other therapeutic foreign proteins.


Subject(s)
GPI-Linked Proteins/antagonists & inhibitors , Immunotoxins/immunology , Lung Neoplasms/therapy , Mesothelioma/therapy , Recombinant Fusion Proteins/immunology , Adaptive Immunity , Animals , Antibodies, Monoclonal/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line, Tumor , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte/genetics , Exotoxins/genetics , Exotoxins/immunology , Female , Humans , Immunoglobulin Variable Region/immunology , Immunotherapy/methods , Immunotoxins/genetics , Immunotoxins/therapeutic use , Lung Neoplasms/immunology , Mesothelin , Mesothelioma/immunology , Mesothelioma, Malignant , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Mutation , Protein Engineering , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Helper-Inducer , Xenograft Model Antitumor Assays
16.
Immunol Rev ; 270(1): 152-64, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26864110

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins designed to treat cancer. They are made up of an Fv or Fab that targets an antigen on a cancer cell fused to a 38-kDa portion of Pseudomonas exotoxin A (PE38). Because PE38 is a bacterial protein, it is highly immunogenic in patients with solid tumors that have normal immune systems, but much less immunogenic in patients with hematologic malignancies where the immune system is suppressed. RITs have shown efficacy in refractory hairy cell leukemia and in some children with acute lymphoblastic leukemia, but have been much less effective in solid tumors, because neutralizing antibodies develop and prevent additional treatment cycles. In this paper we will (i) review data from clinical trials describing the immunogenicity of PE38 in different patient populations; (ii) review results from clinical trials using different immunosuppressive drugs; and (iii) describe our efforts to make new less-immunogenic RITs by identifying and removing T- and B-cell epitopes to hide the RIT from the immune system.


Subject(s)
Immunotoxins/immunology , Immunotoxins/therapeutic use , Recombinant Fusion Proteins , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/immunology , Antibody Formation , Antigens/immunology , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Clinical Trials as Topic , Drug Administration Routes , Drug Therapy, Combination , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Exotoxins/chemistry , Exotoxins/genetics , Exotoxins/immunology , Genetic Engineering , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/therapeutic use , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Immunotherapy , Immunotoxins/administration & dosage , Immunotoxins/adverse effects , Immunotoxins/chemistry , Immunotoxins/genetics , Mesothelin , Mice , Neoplasms/immunology , Neoplasms/therapy , Polyethylene Glycols , Sequence Deletion , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
17.
Mol Cancer Ther ; 14(12): 2789-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26443804

ABSTRACT

SS1P is a recombinant immunotoxin (RIT) that targets mesothelin. It consists of an antimesothelin Fv fused to a portion of Pseudomonas exotoxin A. In clinical studies, it has produced dramatic responses in patients with advanced mesothelioma, when combined with immunosuppressive therapy so that several treatment cycles could be given. Otherwise its activity is limited by its immunogenicity. In this work, we describe the development and characterization of LMB-T20, a highly potent RIT targeted at mesothelin-expressing cancers with low immunogenicity due to removal of its eight T-cell epitopes. LMB-T20 was more active than SS1P when tested on four different mesothelin-expressing cell lines as well as on cells obtained from patients with mesothelioma. It also has potent antitumor activity in mice, and has reduced immunogenicity as measured by cytokine secretion assays. In conclusion, LMB-T20 is a favorable candidate for evaluation in clinical trials due to its reduced immunogenicity and excellent activity.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Epitopes, T-Lymphocyte/immunology , GPI-Linked Proteins/immunology , Mesothelioma/drug therapy , Recombinant Proteins/administration & dosage , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/genetics , Exotoxins/genetics , Exotoxins/immunology , GPI-Linked Proteins/biosynthesis , Humans , Immunogenetic Phenomena/drug effects , Mesothelin , Mesothelioma/genetics , Mesothelioma/immunology , Mice , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Virulence Factors/genetics , Virulence Factors/immunology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
18.
J Immunol Methods ; 425: 10-20, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26056938

ABSTRACT

The ability to identify immunogenic determinants that activate T-cells is important for the development of new vaccines, allergy therapy and protein therapeutics. In silico MHC-II binding prediction algorithms are often used for T-cell epitope identification. To understand how well those programs predict immunogenicity, we computed HLA binding to peptides spanning the sequence of PE38, a fragment of an anti-cancer immunotoxin, and compared the predicted and experimentally identified T-cell epitopes. We found that the prediction for individual donors did not correlate well with the experimental data. Furthermore, prediction of T-cell epitopes in an HLA heterogenic population revealed that the two strongest epitopes were predicted at multiple cutoffs but the third epitope was predicted negative at all cutoffs and overall 4/9 epitopes were missed at several cutoffs. We conclude that MHC class-II binding predictions are not sufficient to predict the T-cell epitopes in PE38 and should be supplemented by experimental work.


Subject(s)
ADP Ribose Transferases/immunology , Bacterial Toxins/immunology , Epitopes, T-Lymphocyte/immunology , Exotoxins/immunology , HLA-DR Antigens/immunology , Lymphocyte Activation/immunology , Protein Binding/immunology , Pseudomonas/immunology , T-Lymphocytes/immunology , Virulence Factors/immunology , Algorithms , Amino Acid Sequence , Antibody Formation/immunology , Binding Sites/immunology , Humans , Molecular Sequence Data , Peptides/immunology , Pseudomonas aeruginosa Exotoxin A
19.
Proc Natl Acad Sci U S A ; 111(23): 8571-6, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24799704

ABSTRACT

Nonhuman proteins have valuable therapeutic properties, but their efficacy is limited by neutralizing antibodies. Recombinant immunotoxins (RITs) are potent anticancer agents that have produced many complete remissions in leukemia, but immunogenicity limits the number of doses that can be given to patients with normal immune systems. Using human cells, we identified eight helper T-cell epitopes in PE38, a portion of the bacterial protein Pseudomonas exotoxin A which consists of the toxin moiety of the RIT, and used this information to make LMB-T18 in which three epitopes were deleted and five others diminished by point mutations in key residues. LMB-T18 has high cytotoxic and antitumor activity and is very resistant to thermal denaturation. The new immunotoxin has a 93% decrease in T-cell epitopes and should have improved efficacy in patients because more treatment cycles can be given. Furthermore, the deimmunized toxin can be used to make RITs targeting other antigens, and the approach we describe can be used to deimmunize other therapeutically useful nonhuman proteins.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunotoxins/immunology , Neoplasms/immunology , Recombinant Fusion Proteins/immunology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Amino Acids/genetics , Amino Acids/immunology , Animals , Antibody Formation/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Electrophoresis, Polyacrylamide Gel , Epitope Mapping , Exotoxins/genetics , Exotoxins/immunology , Female , Humans , Immunotherapy/methods , Immunotoxins/genetics , Immunotoxins/therapeutic use , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, SCID , Models, Molecular , Neoplasms/pathology , Neoplasms/therapy , Peptides/genetics , Peptides/immunology , Point Mutation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Virulence Factors/genetics , Virulence Factors/immunology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
20.
Proc Natl Acad Sci U S A ; 111(23): 8577-82, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24843166

ABSTRACT

Immune responses can make protein therapeutics ineffective or even dangerous. We describe a general computational protein design method for reducing immunogenicity by eliminating known and predicted T-cell epitopes and maximizing the content of human peptide sequences without disrupting protein structure and function. We show that the method recapitulates previous experimental results on immunogenicity reduction, and we use it to disrupt T-cell epitopes in GFP and Pseudomonas exotoxin A without disrupting function.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunotoxins/immunology , Protein Engineering/methods , Proteins/immunology , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cell Line, Tumor , Computer-Aided Design , Epitopes, T-Lymphocyte/genetics , Exotoxins/chemistry , Exotoxins/genetics , Exotoxins/immunology , Flow Cytometry , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Immunization , Immunotoxins/chemistry , Immunotoxins/genetics , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Sequence Homology, Amino Acid , Spectrometry, Fluorescence , Support Vector Machine , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
SELECTION OF CITATIONS
SEARCH DETAIL
...