Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 117(11): 6086-6091, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123080

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins composed of an Fv and a protein toxin being developed for cancer treatment. The Fv brings the toxin to the cancer cell, but most of the RITs do not reach the tumor and are removed by other organs. To identify cells responsible for RIT removal, and the pathway by which RITs reach these cells, we studied SS1P, a 63-kDa RIT that targets mesothelin-expressing tumors and has a short serum half-life. The major organs that remove RIT were identified by live mouse imaging of RIT labeled with FNIR-Z-759. Cells responsible for SS1P removal were identified by immunohistochemistry and intravital two-photon microscopy of kidneys of rats. The primary organ of SS1P removal is kidney followed by liver. In the kidney, SS1P passes through the glomerulus, is taken up by proximal tubular cells, and transferred to lysosomes. In the liver, macrophages are involved in removal. The short half-life of SS1P is due to its very rapid filtration by the kidney followed by degradation in proximal tubular cells of the kidney. In mice treated with SS1P, proximal tubular cells are damaged and albumin in the urine is increased. SS1P uptake by kidney is reduced by coadministration of l-lysine. Our data suggests that l-lysine administration to humans might prevent SS1P-mediated kidney damage, reduce albumin loss in urine, and alleviate capillary leak syndrome.


Subject(s)
Albuminuria/pathology , Antibodies, Monoclonal/pharmacokinetics , Capillary Leak Syndrome/pathology , Immunotoxins/pharmacokinetics , Kidney Tubules, Proximal/drug effects , Albuminuria/chemically induced , Albuminuria/prevention & control , Albuminuria/urine , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/toxicity , Capillary Leak Syndrome/chemically induced , Capillary Leak Syndrome/prevention & control , Capillary Leak Syndrome/urine , Disease Models, Animal , Female , Fluorescent Dyes/chemistry , Half-Life , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , Immunotoxins/toxicity , Intravital Microscopy , Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/diagnostic imaging , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Lysine/administration & dosage , Mesothelin , Mice , Microscopy, Fluorescence , Neoplasms/drug therapy , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/toxicity , Renal Elimination/drug effects , Serum Albumin/analysis , Serum Albumin/metabolism , Staining and Labeling
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.
Proc Natl Acad Sci U S A ; 115(15): E3501-E3508, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581296

ABSTRACT

Recombinant immunotoxins (RITs) are chimeric proteins consisting of a Fv that binds to a cancer cell and a portion of a protein toxin. One of these, Moxetumomab pasudotox, was shown to be effective in treating patients with some leukemias, where the cells are readily accessible to the RIT. However, their short half-life limits their efficacy in solid tumors, because penetration into the tumors is slow. Albumin and agents bound to albumin have a long half-life in the circulation. To increase the time tumor cells are exposed to RITs, we have produced and evaluated variants that contain either an albumin-binding domain (ABD) from Streptococcus or single-domain antibodies from Llama. We have inserted these ABDs into RITs targeting mesothelin, between the Fv and the furin cleavage site. We find that these proteins can be produced in large amounts, are very cytotoxic to mesothelin-expressing cancer cell lines, and have a high affinity for human or mouse serum albumin. In mice, the RIT containing an ABD from Streptococcus has a longer half-life and higher antitumor activity than the other two. Its half-life in the circulation of mice ranges from 113 to 194 min compared with 13 min for an RIT with no ABD. Cell uptake studies show the RIT enters the target cell bound to serum albumin. We conclude that RITs with improved half-lives and antitumor activity should be evaluated for the treatment of cancer in humans.


Subject(s)
Immunotoxins/pharmacokinetics , Animals , Bacterial Toxins/pharmacokinetics , Bacterial Toxins/pharmacology , Cell Line, Tumor/drug effects , Disease Models, Animal , Exotoxins/pharmacokinetics , Exotoxins/pharmacology , Female , GPI-Linked Proteins/drug effects , Half-Life , Humans , Immunotoxins/immunology , Leukemia/drug therapy , Mesothelin , Mice , Mice, Nude , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Serum Albumin/metabolism , Serum Albumin/therapeutic use
4.
Blood ; 130(14): 1620-1627, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28983018

ABSTRACT

Novel therapies are needed to overcome chemotherapy resistance for children with relapsed/refractory acute lymphoblastic leukemia (ALL). Moxetumomab pasudotox is a recombinant anti-CD22 immunotoxin. A multicenter phase 1 study was conducted to determine the maximum-tolerated cumulative dose (MTCD) and evaluate safety, activity, pharmacokinetics, and immunogenicity of moxetumomab pasudotox in children, adolescents, and young adults with ALL (N = 55). Moxetumomab pasudotox was administered as a 30-minute IV infusion at doses of 5 to 50 µg/kg every other day for 6 (cohorts A and B) or 10 (cohort C) doses in 21-day cycles. Cohorts B and C received dexamethasone prophylaxis against capillary leak syndrome (CLS). The most common treatment-related adverse events were reversible weight gain, hepatic transaminase elevation, and hypoalbuminemia. Dose-limiting CLS occurred in 2 of 4 patients receiving 30 µg/kg of moxetumomab pasudotox every other day for 6 doses. Incorporation of dexamethasone prevented further dose-limiting CLS. Six of 14 patients receiving 50 µg/kg of moxetumomab pasudotox for 10 doses developed hemolytic uremic syndrome (HUS), thrombotic microangiopathy (TMA), or HUS-like events, exceeding the MTCD. Treatment expansion at 40 µg/kg for 10 doses (n = 11) exceeded the MTCD because of 2 HUS/TMA/HUS-like events. Dose level 6B (ie, 50 µg/kg × 6 doses) was the MTCD, selected as the recommended phase 2 dose. Among 47 evaluable patients, an objective response rate of 32% was observed, including 11 (23%) composite complete responses, 5 of which were minimal residual disease negative by flow cytometry. Moxetumomab pasudotox showed a manageable safety profile and evidence of activity in relapsed or refractory childhood ALL. This trial was registered at www.clinicaltrials.gov as #NCT00659425.


Subject(s)
Bacterial Toxins/therapeutic use , Exotoxins/therapeutic use , Immunotoxins/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Sialic Acid Binding Ig-like Lectin 2/immunology , Adolescent , Adult , Bacterial Toxins/adverse effects , Bacterial Toxins/immunology , Bacterial Toxins/pharmacokinetics , Capillary Leak Syndrome/prevention & control , Child , Child, Preschool , Dexamethasone/therapeutic use , Exotoxins/adverse effects , Exotoxins/immunology , Exotoxins/pharmacokinetics , Female , Glucocorticoids/therapeutic use , Hemolytic-Uremic Syndrome/chemically induced , Humans , Hypoalbuminemia/chemically induced , Immunotoxins/adverse effects , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Infant , Male , Maximum Tolerated Dose , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Thrombotic Microangiopathies/chemically induced , Weight Gain/drug effects , Young Adult
5.
Cancer Immunol Immunother ; 66(3): 367-378, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27933426

ABSTRACT

Tumor endothelial marker 1 (TEM1) has been identified as a novel surface marker upregulated on the blood vessels and stroma in many solid tumors. We previously isolated a novel single-chain variable fragment (scFv) 78 against TEM1 from a yeast display scFv library. Here, we evaluated the potential applications of scFv78 as a tool for tumor molecular imaging, immunotoxin-based therapy and nanotherapy. Epitope mapping, three-dimensional structure docking and affinity measurements indicated that scFv78 could bind to both human and murine TEM1, with equivalent affinity, at a well-conserved conformational epitope. The rapid internalization of scFv78 and scFv78-labeled nanoparticles was triggered after specific TEM1 binding. The scFv78-saporin immunoconjugate also exerted dose-dependent cytotoxicity with high specificity to TEM1-positive cells in vitro. Finally, specific and sensitive tumor localization of scFv78 was confirmed with optical imaging in a tumor mouse model that has highly endogenous mTEM1 expression in the vasculature. Our data indicated that scFv78, the first fully human anti-TEM1 recombinant antibody, recognizes both human and mouse TEM1 and has unique and favorable features that are advantageous for the development of imaging probes or antibody-toxin conjugates for a large spectrum of human TEM1-positive solid tumors.


Subject(s)
Antigens, CD/immunology , Antigens, Neoplasm/immunology , Immunoglobulin Fragments/immunology , Immunotoxins/immunology , Nanoparticles/administration & dosage , Neoplasm Proteins/immunology , Neoplasms/immunology , Neoplasms/therapy , Amino Acid Sequence , Animals , Antigens, CD/biosynthesis , Epitopes/immunology , Humans , Immunotherapy/methods , Immunotoxins/pharmacokinetics , Mice , Mice, Nude , Molecular Docking Simulation , Nanoparticles/metabolism , Neoplasm Proteins/biosynthesis
6.
Int J Cancer ; 138(4): 1013-23, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26334777

ABSTRACT

Fibroblast activation protein (FAP) is highly expressed in the tumor-associated fibroblasts (TAFs) of most human epithelial cancers. FAP plays a critical role in tumorigenesis and cancer progression, which makes it a promising target for novel anticancer therapy. However, mere abrogation of FAP enzymatic activity by small molecules is not very effective in inhibiting tumor growth. In this study, we have evaluated a novel immune-based approach to specifically deplete FAP-expressing TAFs in a mouse 4T1 metastatic breast cancer model. Depletion of FAP-positive stromal cells by FAP-targeting immunotoxin αFAP-PE38 altered levels of various growth factors, cytokines, chemokines and matrix metalloproteinases, decreased the recruitment of tumor-infiltrating immune cells in the tumor microenvironment and suppressed tumor growth. In addition, combined treatment with αFAP-PE38 and paclitaxel potently inhibited tumor growth in vivo. Our findings highlight the potential use of immunotoxin αFAP-PE38 to deplete FAP-expressing TAFs and thus provide a rationale for the use of this immunotoxin in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Fibroblasts/metabolism , Gelatinases/antagonists & inhibitors , Immunotoxins/pharmacology , Membrane Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , BALB 3T3 Cells , Disease Models, Animal , Endopeptidases , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Immunotoxins/pharmacokinetics , Mice , Real-Time Polymerase Chain Reaction , Serine Endopeptidases
7.
Proc Natl Acad Sci U S A ; 109(29): 11782-7, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22753489

ABSTRACT

Recombinant immunotoxins (RITs) are hybrid proteins used to treat cancer. These proteins are composed of an Fv that reacts with cancer cells joined to a portion of Pseudomonas exotoxin A, which kills the cell. Because the toxin is a foreign protein, it can induce neutralizing antibodies and thereby limit the number of doses a patient can receive. We previously identified seven major mouse B-cell epitopes in the toxin, and subsequently silenced them using point mutations that converted large hydrophilic amino acids to alanine, yet retained full antitumor activity. Here we present results in which we identify and silence human B-cell epitopes in the RIT HA22. We obtained B cells from patients with antibodies to RITs, isolated the corresponding variable fragments (Fvs), and constructed a phage-display library containing Fvs that bind to the RITs. We then used alanine scanning mutagenesis to locate the epitopes. We found that human and mouse epitopes frequently overlap but are not identical. Most mutations that remove mouse epitopes did not remove human epitopes. Using the epitope information, we constructed a variant immunotoxin, HA22-LR-LO10, which has low reactivity with human antisera, yet has high cytotoxic and antitumor activity and can be given to mice at high doses without excess toxicity. The toxin portion of this RIT (LR-LO10) can be used with Fvs targeting other cancer antigens and is suitable for clinical development.


Subject(s)
Antineoplastic Agents/immunology , Epitopes, B-Lymphocyte/immunology , Immunotherapy/methods , Immunotoxins/immunology , Models, Molecular , Neoplasms/drug therapy , Recombinant Proteins/immunology , Amino Acid Sequence , Animals , Enzyme-Linked Immunosorbent Assay , Humans , Immunotoxins/chemistry , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Mice , Molecular Sequence Data , Mutagenesis , Neoplasms/immunology , Peptide Library , Protein Engineering/methods , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Single-Chain Antibodies/immunology , Statistics, Nonparametric
8.
Int J Cancer ; 135(6): 1497-508, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24523193

ABSTRACT

CMML (chronic myelomonocytic leukemia) belongs to the group of myeloid neoplasms known as myelodysplastic and myeloproliferative diseases. In some patients with a history of CMML, the disease transforms to acute myelomonocytic leukemia (AMML). There are no specific treatment options for patients suffering from CMML except for supportive care and DNA methyltransferase inhibitors in patients with advanced disease. New treatment strategies are urgently required, so we have investigated the use of immunotherapeutic directed cytolytic fusion proteins (CFPs), which are chimeric proteins comprising a selective domain and a toxic component (preferably of human origin to avoid immunogenicity). The human serine protease granzyme B is a prominent candidate for tumor immunotherapy because it is expressed in cytotoxic T lymphocytes and natural killer cells. Here, we report the use of CD64 as a novel target for specific CMML and AMML therapy, and correlate CD64 expression with typical surface markers representing these diseases. We demonstrate that CD64-specific human CFPs kill CMML and AMML cells ex vivo, and that the mutant granzyme B protein R201K is more cytotoxic than the wild-type enzyme in the presence of the granzyme B inhibitor PI9. Besides, the human CFP based on the granzyme B mutant was also able to kill AMML or CMML probes resistant to Pseudomonas exotoxin A.


Subject(s)
Granzymes/administration & dosage , Leukemia, Myelomonocytic, Acute/drug therapy , Leukemia, Myelomonocytic, Chronic/drug therapy , Aged , Aged, 80 and over , Drug Stability , Female , HL-60 Cells , Humans , Immunotoxins/administration & dosage , Immunotoxins/blood , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Leukemia, Myelomonocytic, Acute/immunology , Leukemia, Myelomonocytic, Chronic/immunology , Male , Middle Aged , Receptors, IgG/biosynthesis , Receptors, IgG/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics
9.
Bioconjug Chem ; 24(5): 772-9, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23578050

ABSTRACT

Antibody-drug conjugates (ADCs) are target-specific anticancer agents consisting of cytotoxic drugs covalently linked to a monoclonal antibody. The number of ADCs in the clinic is growing, and therefore thorough characterization of the quantitative assays used to measure ADC concentrations in support of pharmacokinetic, efficacy, and safety studies is of increasing importance. Cytotoxic drugs such as the tubulin polymerization inhibiting auristatin, monomethyl auristatin E, have been conjugated to antibodies via cleavable linkers (MC-vc-PAB) through internal cysteines. This results in a heterogeneous mixture of antibody species with drug-to-antibody ratios (DAR) ranging from 0 to 8. In order to characterize the assays used to quantitate total MC-vc-PAB-MMAE ADCs (conjugated and unconjugated antibody), we used purified fractions with defined DARs from 6 therapeutic antibodies to evaluate different assay formats and reagents. Our investigations revealed that for quantitation of total antibody, including all unconjugated and conjugated antibody species, sandwich ELISA formats did not always allow for recovery of all purified DAR fractions (DAR 0-8) to within ±20% of the expected values at the reagent concentrations tested. In evaluating alternative approaches, we found that the recovery of DAR fractions with semihomogeneous assay (SHA) formats, in which sample, capture, and detection reagents are preincubated in solution, were less affected by the antibody's MMAE drug load as compared to traditional stepwise sandwich ELISAs. Thus, choosing the optimal assay format and reagents for total antibody assays is valuable for developing accurate quantitative assays.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Immunotoxins/pharmacokinetics , Oligopeptides/pharmacokinetics , Tubulin Modulators/pharmacokinetics , Animals , Antineoplastic Agents/chemistry , Enzyme-Linked Immunosorbent Assay , Immunotoxins/chemistry , Mice , Mice, SCID , Oligopeptides/chemistry , Tubulin Modulators/chemistry
10.
Future Oncol ; 9(3): 355-68, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23469971

ABSTRACT

Antibody-drug conjugates (ADCs) are a broad class of molecules comprising of a potent cytotoxic agent conjugated with a monoclonal antibody using a chemically stable linker. By selecting a monoclonal antibody directed against a tumor-specific or tumor-associated antigen, ADCs allow the targeted delivery of highly potent cytotoxic agents that result in unacceptable toxicity when administered as free agents. ADCs are currently being developed for the treatment of a wide variety of tumors. In this review, the current clinical and preclinical status of ADCs for the treatment of B-cell non-Hodgkin's lymphoma and B-cell leukemia will be discussed. ADCs have the potential to alter treatment paradigms for these diseases by providing both increased efficacy and improved safety and tolerability over current chemotherapy-based regimens.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Leukemia/drug therapy , Lymphoma, B-Cell/drug therapy , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antineoplastic Agents/pharmacokinetics , Clinical Trials as Topic , Drug Carriers , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Inotuzumab Ozogamicin , Leukemia/metabolism , Lymphoma, B-Cell/metabolism , Maytansine/analogs & derivatives , Maytansine/therapeutic use
11.
Xenobiotica ; 43(1): 84-97, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23199366

ABSTRACT

1.When the immune system encounters incoming infectious agents, this generally leads to immunity. The evoked immune response is usually robust, but can be severely perturbed by potentially harmful environmental agents such as chemicals, pharmaceuticals and allergens. 2.Immunosuppression, hypersensitivity and autoimmunity may occur due to changed immune activity. Evaluation of the immunotoxic potency of agents as part of risk assessment is currently established in vivo with animal models and in vitro with cell lines or primary cells. 3.Although in vivo testing is usually the most relevant situation for many agents, more and more in vitro models are being developed for assessment of immunotoxicity. In this context, hypersensitivity and immunosuppression are considered to be a primary focus for developing in vitro methods. Three-dimensional organotypic tissue models are also part of current research in immunotoxicology. 4.In recent years, there has been a revival of interest in organotypic tissue models. In the context of immunotoxicity testing, precision-cut lung slices in particular have been intensively studied. Therefore, this review is very much focused on pulmonary immunotoxicology. Respiratory hypersensitivity and inflammation are further highlighted aspects of this review. Immunotoxicity assessment currently is of limited use in other tissue models, which are therefore described only briefly within this review.


Subject(s)
Immunotoxins , Lung/metabolism , Models, Biological , Tissue Culture Techniques/methods , Xenobiotics , Allergens/immunology , Allergens/pharmacokinetics , Allergens/pharmacology , Animals , Autoimmunity/drug effects , Humans , Immune Tolerance/drug effects , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , Lung/pathology , Xenobiotics/immunology , Xenobiotics/pharmacokinetics , Xenobiotics/pharmacology
12.
Mol Imaging ; 11(5): 408-16, 2012.
Article in English | MEDLINE | ID: mdl-22954185

ABSTRACT

Epidermal growth factor receptor (EGFR) signaling inhibition represents a highly promising arena for the application of molecularly targeted cancer therapies. EGFR conjugated metal chelates have been proposed as potential imaging agents for cancers that overexpress EGFR receptors. Through improved understanding of EGFR biology in human cancers, there is anticipation that more tumor-selective therapy approaches with diminished collateral normal tissue toxicity can be advanced. We report here on the results with a thermodynamically stable chelate, 1,4,7-tris(carboxymethyl)-10-(2-aminoethyl)-1,4,7,10-tetraazacyclododecane (DO3A-EA) and anti-EGFr (ior egf/r3) conjugate to develop immunospecific imaging agent. Conjugation and labelling with anti-EGFr was performed using standard procedure and subjected to purification on size exclusion chromatography. The conjugated antibodies were labeled with a specific activity 20-30 mCi/mg of protein. Labeling efficiencies were measured by ascending paper chromatography on ITLC-SG strips. Radiolabeling of the immunoconjugate was found to be 98.5 ± 0.30%. (99m)Tc-DO3A-EA-EGFr conjugate was studied in athymic mice bearing U-87MG, MDA-MB-468 tumors following intravenous injection. Pharmacokinetic and biodistribution studies confirmed long circulation times (t(1/2)(fast)  =  45 min and t1/2(slow)  = 4 hours 40 min) and efficient accumulation in tumors. Biodistribution studies in athymic mice grafted with U-87MG human glioblastoma multiforme and Hela human cervical carcinoma tumors revealed significant localization of (99m)Tc-labeled antibodies conjugate in tumors and reduced accumulation in normal organs. This new chelating agent is promising for immunoscintigraphy since good tumour-to-normal organ contrast could be demonstrated. These properties can be exploited for immunospecifc contrast agents in nuclear medicine and SPECT imaging.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , ErbB Receptors/antagonists & inhibitors , Glioma/drug therapy , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Immunotoxins/pharmacokinetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Chelating Agents/chemistry , Contrast Media/chemistry , Drug Delivery Systems/methods , Drug Stability , ErbB Receptors/immunology , Glioma/immunology , Glioma/metabolism , HeLa Cells , Heterocyclic Compounds, 1-Ring/administration & dosage , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Immunotoxins/administration & dosage , Immunotoxins/chemistry , Immunotoxins/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Imaging/methods , Molecular Targeted Therapy , Rabbits , Radionuclide Imaging , Technetium , Whole Body Imaging , Xenograft Model Antitumor Assays
13.
Cancer ; 118(23): 5733-40, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22648179

ABSTRACT

BACKGROUND: We conducted a phase 1, multicenter, open-label, dose-escalation study (TDM3569g) to assess the safety, tolerability, and pharmacokinetics of single-agent trastuzumab emtansine (T-DM1) administered weekly and once every 3 weeks in patients with HER2-positive metastatic breast cancer previously treated with trastuzumab. The weekly dose results are described here. METHODS: Patients were administered escalating doses of T-DM1 weekly, starting at 1.2 mg/kg. Additional patients were enrolled at the maximum tolerated dose (MTD) to better characterize tolerability and pharmacokinetics. RESULTS: Twenty-eight patients received weekly T-DM1, and the MTD was determined to be 2.4 mg/kg. In general, T-DM1 was well tolerated, requiring few dose modifications or discontinuations because of adverse events (AEs). Grade ≥ 3 AEs were reported in 19 patients (67.9%); treatment-related AEs occurred in 25 (89.3%) patients. Exposure to weekly T-DM1 was dose-proportional at ≥ 1.2 mg/kg, and accumulation of T-DM1 and total trastuzumab was observed. Objective partial tumor responses were reported in 13 (46.4%) patients; the median duration of response was 18.6 months, and the 6-month clinical benefit rate was 57.1%. CONCLUSION: The results suggest that a weekly dose of T-DM1 2.4 mg/kg has antitumor activity and is well tolerated in patients with HER2-positive metastatic breast cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Maytansine/analogs & derivatives , Receptor, ErbB-2/analysis , Ado-Trastuzumab Emtansine , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/chemistry , Female , Humans , Immunotoxins/administration & dosage , Immunotoxins/adverse effects , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Maximum Tolerated Dose , Maytansine/administration & dosage , Maytansine/adverse effects , Maytansine/immunology , Maytansine/pharmacokinetics , Middle Aged , Trastuzumab
14.
Bioorg Med Chem ; 20(24): 6929-39, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23159039

ABSTRACT

A major drawback of internalizing monoclonal antibodies (mAbs) radioiodinated with direct electrophilic approaches is that tumor retention of radioactivity is compromised by the rapid washout of iodo-tyrosine, the primary labeled catabolite for mAbs labeled via this strategy. In our continuing efforts to develop more versatile residualizing labels that could overcome this problem, we have designed SIB-DOTA, a prosthetic labeling template that combines the features of the prototypical, dehalogenation-resistant N-succinimidyl 3-iodobenzoate (SIB) with DOTA, a useful macrocyclic chelator for labeling with radiometals. Herein we describe the synthesis of the unlabeled standard of this prosthetic moiety, its protected tin precursor, and radioiodinated SIB-DOTA. An anti-EGFRvIII-reactive mAb, L8A4 was radiolabeled with [(131)I]SIB-DOTA in 27.1±6.2% (n=2) conjugation yields and its targeting properties to the same mAb labeled with [(125)I]SGMIB both in vitro and in vivo using U87MG·ΔEGFR cells and xenografts were compared. In vitro paired-label internalization assays showed that the intracellular radioactivity from [(131)I]SIB-DOTA-L8A4 was 21.4±0.5% and 26.2±1.1% of initially bound radioactivity at 16 and 24h, respectively. In comparison, these values for [(125)I]SGMIB-L8A4 were 16.7±0.5% and 14.9±1.1%. Similarly, the SIB-DOTA prosthetic group provided better tumor targeting in vivo than SGMIB over 8 d period. These results suggest that SIB-DOTA warrants further evaluation as a residualizing agent for labeling internalizing mAbs including those targeted to EGFRvIII.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Heterocyclic Compounds, 1-Ring/chemistry , Immunotoxins/chemistry , Immunotoxins/pharmacokinetics , Iodobenzoates/chemistry , Radiopharmaceuticals/chemical synthesis , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , ErbB Receptors/immunology , Glioblastoma/immunology , Glioblastoma/metabolism , Heterocyclic Compounds, 1-Ring/chemical synthesis , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Iodine Radioisotopes/chemistry , Iodine Radioisotopes/pharmacokinetics , Iodobenzoates/chemical synthesis , Iodobenzoates/pharmacokinetics , Isotope Labeling/methods , Mice , Mice, Inbred BALB C , Organometallic Compounds/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/immunology , Radiopharmaceuticals/pharmacokinetics , Tin/chemistry , Tissue Distribution
15.
J Oncol Pharm Pract ; 18(4): 417-20, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22228555

ABSTRACT

Radioimmunotherapy has emerged as a treatment modality for patients with CD20 positive B-cell non-Hodgkin's lymphoma (NHL). Prior to administration of a therapeutic dose, confirmation of uptake of the radiolabeled compound in tumor locations and calculation of an appropriate dose can be performed using a diagnostic dose and subsequent imaging. We report the case of a 69-year-old male with a relapsed mantle cell lymphoma scheduled for radioimmunotherapy, where diagnostic imaging with 131-I labeled rituximab revealed unexpected new insights with implications for treatment. Persistence of the mantle cell lymphoma in a lymph node in the left arm was demonstrated by an 18-F fluorodeoxyglucose scan. However, a scan after a diagnostic dose of 131-I labeled rituximab did not show any uptake of the tracer, even though subsequent cytological analysis unequivocally confirmed a CD20 positive B-cell population in the lesion. The administration of a therapeutic dose of 131-I labeled rituximab was therefore cancelled. We here discuss the mechanisms that may explain lack of targeting in a proven CD20-positive lymphoma and provide recommendations for further studies.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacokinetics , Antigens, CD20/metabolism , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/radiotherapy , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , B-Lymphocytes/metabolism , Humans , Immunotoxins/administration & dosage , Immunotoxins/pharmacokinetics , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/pharmacokinetics , Male , Radioimmunotherapy/methods , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Rituximab
16.
Int J Mol Sci ; 13(12): 16020-45, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23443108

ABSTRACT

Drug conjugates have been studied extensively in preclinical in vitro and in vivo models but to date only a few compounds have progressed to the clinical setting. This situation is now changing with the publication of studies demonstrating a significant impact on clinical practice and highlighting the potential of this new class of targeted therapies. This review summarizes the pharmacological and molecular background of the main drug conjugation systems, namely antibody drug conjugates (ADCs), immunotoxins and immunoliposomes. All these compounds combine the specific targeting moiety of an antibody or similar construct with the efficacy of a toxic drug. The aim of this strategy is to target tumor cells specifically while sparing normal tissue, thus resulting in high efficacy and low toxicity. Recently, several strategies have been investigated in phase I clinical trials and some have entered phase III clinical development. This review provides a detailed overview of various strategies and critically discusses the most relevant achievements. Examples of the most advanced compounds include T-DM1 and brentuximab vedotin. However, additional promising strategies such as immunotoxins and immunoliposmes are already in clinical development. In summary, targeted drug delivery by drug conjugates is a new emerging class of anti-cancer therapy that may play a major role in the future.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents , Immunoconjugates , Immunotoxins , Liposomes , Maytansine/analogs & derivatives , Neoplasms , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Brentuximab Vedotin , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Liposomes/immunology , Liposomes/pharmacokinetics , Liposomes/therapeutic use , Maytansine/immunology , Maytansine/pharmacokinetics , Maytansine/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Trastuzumab
17.
Life Sci ; 289: 120222, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34902436

ABSTRACT

In order to overcome limitations of conventional cancer therapy methods, immunotoxins with the capability of target-specific action have been designed and evaluated pre-clinically, and some of them are in clinical studies. Targeting cancer cells via antibodies specific for tumour-associated surface proteins is a new biomedical approach that could provide the selectivity that is lacking in conventional cancer therapy methods such as radiotherapy and chemotherapy. A successful example of an approved immunotoxin is represented by immunoRNases. ImmunoRNases are fusion proteins in which the toxin has been replaced by a ribonuclease. Conjugation of RNase molecule to monoclonal antibody or antibody fragment was shown to enhance specific cell-killing by several orders of magnitude, both in vitro and in animal models. There are several RNases obtained from different mammalian cells that are expected to be less immunogenic and systemically toxic. In fact, RNases are pro-toxins which become toxic only upon their internalization in target cells mediated by the antibody moiety. The structure and large size of the antibody molecules assembled with the immunoRNases have always been a challenge in the application of immunoRNases as an antitoxin. To overcome this obstacle, we have offered a new strategy for the application of immunoRNases as a promising approach for upgrading immunoRNAses with maximum affinity and high stability in the cell, which can ultimately act as an effective large-scale cancer treatment. In this review, we introduce the optimized antibody-like molecules with small size, approximately 10 kD, which are presumed to significantly enhance RNase activity and be a suitable agent with the potential for anti-cancer functionality. In addition, we also discuss new molecular entities such as monobody, anticalin, nonobody and affilin as refined versions in the development of immunoRNases. These small molecules express their functionality with the suitable small size as well as with low immunogenicity in the cell, as a part of immunoRNases.


Subject(s)
Antineoplastic Agents, Immunological , Antineoplastic Agents , Immunotoxins , Neoplasms , Recombinant Fusion Proteins , Ribonucleases , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , Neoplasms/drug therapy , Neoplasms/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Ribonucleases/genetics , Ribonucleases/immunology , Ribonucleases/pharmacokinetics , Ribonucleases/pharmacology
18.
Cancer Sci ; 102(7): 1396-402, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21481097

ABSTRACT

Fibrin clots in non-malignant conditions form only at the onset or during the active stage of disease and disappear within a few weeks as a result of plasmin digestion or replacement with collagen. In contrast, fibrin clot formation and subsequent replacement with collagen in cancer persist for as long as the cancer cells survive in the body. We developed an anti-fibrin chimeric antibody that reacts with fibrin only, and not fibrinogen (the precursor of fibrin), and then attached an anticancer agent (ACA) to the antibody. Thus, the immunoconjugate did not create an immune complex in the blood stream and was selectively accumulated to fibrin clots in the tumor stroma to create a scaffold, from which effective sustained release of the ACA occurred. In a mouse model, the ACA diffused throughout the tumor tissue to damage both tumor cells and vessels, resulting in potent antitumor activity in stroma-rich spontaneous tumors. This new cancer stroma-targeting therapy may result in an increased duration of drug exposure and be a highly effective new therapy, particularly for refractory, stroma-rich cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Fibrin/antagonists & inhibitors , Immunotoxins/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Drug Design , Female , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Irinotecan , Mice , Neoplasms, Experimental/drug therapy
19.
Blood ; 113(16): 3792-800, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-18988862

ABSTRACT

Immunotoxins based on Pseudomonas exotoxin A (PE) are promising anticancer agents that combine a variable fragment (Fv) from an antibody to a tumor-associated antigen with a 38-kDa fragment of PE (PE38). The intoxication pathway of PE immunotoxins involves receptor-mediated internalization and trafficking through endosomes/lysosomes, during which the immunotoxin undergoes important proteolytic processing steps but must otherwise remain intact for eventual transport to the cytosol. We have investigated the proteolytic susceptibility of PE38 immunotoxins to lysosomal proteases and found that cleavage clusters within a limited segment of PE38. We subsequently generated mutants containing deletions in this region using HA22, an anti-CD22 Fv-PE38 immunotoxin currently undergoing clinical trials for B-cell malignancies. One mutant, HA22-LR, lacks all identified cleavage sites, is resistant to lysosomal degradation, and retains excellent biologic activity. HA22-LR killed chronic lymphocytic leukemia cells more potently and uniformly than HA22, suggesting that lysosomal protease digestion may limit immunotoxin efficacy unless the susceptible domain is eliminated. Remarkably, mice tolerated doses of HA22-LR at least 10-fold higher than lethal doses of HA22, and these higher doses exhibited markedly enhanced antitumor activity. We conclude that HA22-LR advances the therapeutic efficacy of HA22 by using an approach that may be applicable to other PE-based immunotoxins.


Subject(s)
ADP Ribose Transferases/pharmacology , Antibodies, Monoclonal/pharmacology , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Immunoglobulin Variable Region/pharmacology , Immunotoxins/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Sialic Acid Binding Ig-like Lectin 2 , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , ADP Ribose Transferases/adverse effects , ADP Ribose Transferases/genetics , ADP Ribose Transferases/pharmacokinetics , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacokinetics , Bacterial Toxins/adverse effects , Bacterial Toxins/genetics , Bacterial Toxins/pharmacokinetics , Clinical Trials as Topic , Endosomes/metabolism , Exotoxins/adverse effects , Exotoxins/genetics , Exotoxins/pharmacokinetics , Female , Humans , Immunoglobulin Variable Region/adverse effects , Immunoglobulin Variable Region/genetics , Immunotoxins/adverse effects , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lysosomes/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Virulence Factors/adverse effects , Virulence Factors/genetics , Virulence Factors/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
20.
Lasers Surg Med ; 43(7): 721-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22057500

ABSTRACT

Photochemical internalization (PCI) is a method for intracellular delivery of hydrophilic macromolecular drugs with intracellular targets as well as other drugs with limited ability to penetrate cellular membranes. Such drugs enter cells by means of endocytosis and are to a large extent degraded by hydrolytic enzymes in the lysosomes unless they possess a mechanism for cytosolic translocation. PCI is based on photodynamic therapy (PDT) specifically targeting the endosomes and lysosomes of the cells, so that the drugs in these vesicles can escape into the cytosol from where they can reach their targets. The preferential retention of the photosensitizer (PS) in tumor tissue in combination with controlled light delivery makes PCI relatively selective for cancer tissue. The tumor specificity of PCI can be further increased by delivery of drugs that selectively target the tumors. Indeed, this has been shown by PCI delivery of several targeted protein toxins. Targeted protein toxins may be regarded as ideal drugs for PCI delivery, and may represent the clinical future for the PCI technology.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunotoxins/administration & dosage , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Photochemotherapy , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Endosomes/drug effects , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/therapeutic use , Lysosomes/drug effects , Macromolecular Substances/administration & dosage , Macromolecular Substances/pharmacokinetics , Macromolecular Substances/therapeutic use , Photosensitizing Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL