Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 327
Filter
1.
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 ImmunoRNases are fusion proteins in which the toxin has been replaced by a ribonuclease.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
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.
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
4.
Mol Cancer Ther ; 19(7): 1385-1394, 2020 07.
Article in English | MEDLINE | ID: mdl-32398269

ABSTRACT

The EphA2 receptor is found at high levels in tumors and low levels in normal tissue and high EphA2 expression in biopsies is a predictor of poor outcome in patients. Drug discovery groups have therefore sought to develop EphA2-based therapies using small molecule, peptide, and nanoparticle-based approaches (1-3). However, until now only EphA2-targeting antibody-drug conjugates (ADC) have entered clinical development. For example, MEDI-547 is an EphA2-targeting ADC that displayed encouraging antitumor activity in preclinical models and progressed to phase I clinical testing in man. Here we describe the development of BT5528, a bicyclic peptide ("Bicycle") conjugated to the auristatin derivative maleimidocaproyl-monomethyl auristatin E to generate the Bicycle toxin conjugate BT5528. The report compares and contrasts the Pharmacokinetics (PK) characteristics of antibody and Bicycle-based targeting systems and discusses how the PK and payload characteristics of different delivery systems impact the efficacy-toxicity trade off which is key to the development of successful cancer therapies. We show that BT5528 gives rise to rapid update into tumors and fast renal elimination followed by persistent toxin levels in tumors without prolonged exposure of parent drug in the vasculature. This fast in, fast out kinetics gave rise to more favorable toxicology findings in rats and monkeys than were observed with MEDI-547 in preclinical and clinical studies.Graphical Abstract: http://mct.aacrjournals.org/content/molcanther/19/7/1385/F1.large.jpg.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Fibrosarcoma/drug therapy , Oligopeptides/chemistry , Peptides, Cyclic/pharmacology , Receptor, EphA2/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacokinetics , Apoptosis , Cell Proliferation , Female , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Immunotoxins/pharmacokinetics , Immunotoxins/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/administration & dosage , Peptides, Cyclic/pharmacokinetics , Receptor, EphA2/genetics , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
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
6.
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
7.
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
8.
J Cancer Res Clin Oncol ; 143(10): 2025-2038, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28667390

ABSTRACT

PURPOSE: Despite great progress in the diagnosis and treatment of localized prostate cancer (PCa), there remains a need for new diagnostic markers that can accurately distinguish indolent and aggressive variants. One promising approach is the antibody-based targeting of prostate stem cell antigen (PSCA), which is frequently overexpressed in PCa. Here, we show the construction of a molecular imaging probe comprising a humanized scFv fragment recognizing PSCA genetically fused to an engineered version of the human DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT), the SNAP-tag, enabling specific covalent coupling to various fluorophores for diagnosis of PCa. Furthermore, the recombinant immunotoxin (IT) PSCA(scFv)-ETA' comprising the PSCA(scFv) and a truncated version of Pseudomonas exotoxin A (PE, ETA') was generated. METHODS: We analyzed the specific binding and internalization behavior of the molecular imaging probe PSCA(scFv)-SNAP in vitro by flow cytometry and live cell imaging, compared to the corresponding IT PSCA(scFv)-ETA'. The cytotoxic activity of PSCA(scFv)-ETA' was tested using cell viability assays. Specific binding was confirmed on formalin-fixed paraffin-embedded tissue specimen of early and advanced PCa. RESULTS: Alexa Fluor® 647 labeling of PSCA(scFv)-SNAP confirmed selective binding to PSCA, leading to rapid internalization into the target cells. The recombinant IT PSCA(scFv)-ETA' showed selective binding leading to internalization and efficient elimination of target cells. CONCLUSIONS: Our data demonstrate, for the first time, the specific binding, internalization, and cytotoxicity of a scFv-based fusion protein targeting PSCA. Immunohistochemical staining confirmed the specific ex vivo binding to primary PCa material.


Subject(s)
ADP Ribose Transferases/administration & dosage , Antigens, Neoplasm/immunology , Bacterial Toxins/administration & dosage , Exotoxins/administration & dosage , Immunotoxins/administration & dosage , Neoplasm Proteins/immunology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/therapy , Recombinant Fusion Proteins/administration & dosage , Virulence Factors/administration & dosage , ADP Ribose Transferases/immunology , Bacterial Toxins/immunology , Exotoxins/immunology , Flow Cytometry , GPI-Linked Proteins/immunology , HEK293 Cells , Humans , Immunologic Tests , Immunotherapy/methods , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Male , Molecular Targeted Therapy , Prostatic Neoplasms/immunology , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/immunology , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
9.
Cancer Biother Radiopharm ; 32(3): 83-89, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28380302

ABSTRACT

OBJECTIVES: Biodistribution of antibodies is vital to successful immunoscintigraphy/immunotherapy, and it is assumed to be similar to antigen distribution. We measured and compared the binding pattern of radiolabeled antibody to tissue antigen distribution in a nude mouse xenograft model of human melanoma. METHODS: We transplanted 107 FEM-XII human melanoma cells into the right flank of five nude mice. For the control, we transplanted 5 × 106 LS174T human colon cancer cells into the left flank. Two weeks later, 10 µCi of 131I-labeled melanoma-associated 96.5 monoclonal antibody (targeting p97 antigen) was intravenously injected. Three days later, we sacrificed the mice and evaluated 96.5 antibody binding and concentration in the tumors by ex vivo quantitative autoradiography (QAR). Two months later, we incubated adjacent tumor tissue slices in various concentrations of 125I-labeled 96.5 MoAb and evaluated the distribution/concentration of p97 antigen by in vitro QAR. RESULTS: p97 antigen distribution was homogeneous in the tumors (total antigen concentration [Bmax] = 17.36-38.36 pmol/g). In contrast, radiolabeled 96.5 antibody binding was heterogenous between location within the tumor (estimated bound antigen concentration = 0.7-6.6 pmol/g). No quantifiable parameters were found to be related with radiolabeled antibody binding and tumor antigen distribution. Antibody-bound tumor antigen to total antigen ratios ranged between 2% and 38%. CONCLUSIONS: Heterogeneous features of target antibody binding were observed in contrast to relatively homogenous feature of tumor antigen. We did not identify any correlations between p97 antigen distribution and 96.5 antibody binding in melanoma tissue. Radiolabeled 96.5 antibody binding patterns within melanoma cannot be predicted based on p97 antigen distribution in the tumor, which needs to be further studied with several other methods and more subjects in the future.


Subject(s)
Antibodies, Monoclonal/immunology , Immunotoxins/pharmacokinetics , Melanoma-Specific Antigens/immunology , Melanoma/immunology , Animals , Antigen-Antibody Reactions , Humans , Immunotoxins/immunology , Immunotoxins/pharmacology , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/pharmacokinetics , Melanoma/radiotherapy , Mice , Mice, Nude , Tissue Distribution , Xenograft Model Antitumor Assays
10.
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
11.
AAPS J ; 18(5): 1117-1130, 2016 09.
Article in English | MEDLINE | ID: mdl-27287046

ABSTRACT

Antibody-drug conjugates exhibit complex pharmacokinetics due to their combination of macromolecular and small molecule properties. These issues range from systemic concerns, such as deconjugation of the small molecule drug during the long antibody circulation time or rapid clearance from nonspecific interactions, to local tumor tissue heterogeneity, cell bystander effects, and endosomal escape. Mathematical models can be used to study the impact of these processes on overall distribution in an efficient manner, and several types of models have been used to analyze varying aspects of antibody distribution including physiologically based pharmacokinetic (PBPK) models and tissue-level simulations. However, these processes are quantitative in nature and cannot be handled qualitatively in isolation. For example, free antibody from deconjugation of the small molecule will impact the distribution of conjugated antibodies within the tumor. To incorporate these effects into a unified framework, we have coupled the systemic and organ-level distribution of a PBPK model with the tissue-level detail of a distributed parameter tumor model. We used this mathematical model to analyze new experimental results on the distribution of the clinical antibody-drug conjugate Kadcyla in HER2-positive mouse xenografts. This model is able to capture the impact of the drug-antibody ratio (DAR) on tumor penetration, the net result of drug deconjugation, and the effect of using unconjugated antibody to drive ADC penetration deeper into the tumor tissue. This modeling approach will provide quantitative and mechanistic support to experimental studies trying to parse the impact of multiple mechanisms of action for these complex drugs.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Immunotoxins/pharmacokinetics , Maytansine/analogs & derivatives , Models, Theoretical , Tissue Distribution/physiology , Trastuzumab/pharmacokinetics , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Dose-Response Relationship, Drug , Immunotoxins/administration & dosage , Maytansine/administration & dosage , Maytansine/pharmacokinetics , Mice , Mice, Nude , Tissue Distribution/drug effects , Trastuzumab/administration & dosage , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/physiology , Xenograft Model Antitumor Assays/methods
12.
Cancer Lett ; 372(2): 201-9, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-26806809

ABSTRACT

Triple-negative breast cancer (TNBC) is associated with poor prognosis and high prevalence among young premenopausal women. Unlike in other breast cancer subtypes, no targeted therapy is currently available. Overexpression of epithelial cell adhesion molecule (EpCAM) in 60% of TNBC tumors correlates with poorer prognosis and is associated with cancer stem cell phenotype. Thus, selective elimination of EpCAM(+) TNBC tumor cells is of clinical importance. Therefore, we constructed a fully human targeted cytolytic fusion protein, designated GbR201K-αEpCAM(scFv), in which an EpCAM-selective single-chain antibody fragment (scFv) is genetically fused to a granzyme B (Gb) mutant with reduced sensitivity to its natural inhibitor serpin B9. In vitro studies confirmed its specific binding, internalization and cytotoxicity toward a panel of EpCAM-expressing TNBC cells. Biodistribution kinetics and tumor-targeting efficacy using MDA-MB-468 cells in a human TNBC xenograft model in mice revealed selective accumulation of GbR201K-αEpCAM(scFv) in the tumors after i.v. injection. Moreover, treatment of tumor-bearing mice demonstrated a prominent inhibition of tumor growth of up to 50 % in this proof-of-concept study. Taken together, our results indicate that GbR201K-αEpCAM(scFv) is a promising novel targeted therapeutic for the treatment of TNBC.


Subject(s)
Apoptosis/drug effects , Cell Adhesion Molecules/antagonists & inhibitors , Granzymes/pharmacology , Immunotherapy/methods , Immunotoxins/pharmacology , Single-Chain Antibodies/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Tumor Burden/drug effects , Animals , Antibody Specificity , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cell Adhesion Molecule , Female , Granzymes/genetics , Granzymes/pharmacokinetics , HEK293 Cells , Humans , Immunotoxins/genetics , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Mice, Inbred BALB C , Mice, Nude , Mutation , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution , Transfection , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
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
14.
Mol Cancer Ther ; 14(5): 1130-40, 2015 May.
Article in English | MEDLINE | ID: mdl-25724665

ABSTRACT

Antibody-drug conjugates (ADC) are emerging as powerful cancer treatments that combine antibody-mediated tumor targeting with the potent cytotoxic activity of toxins. We recently reported the development of a novel ADC that delivers the cytotoxic payload monomethyl auristatin E (MMAE) to tumor cells expressing tissue factor (TF). By carefully selecting a TF-specific antibody that interferes with TF:FVIIa-dependent intracellular signaling, but not with the procoagulant activity of TF, an ADC was developed (TF-011-MMAE/HuMax-TF-ADC) that efficiently kills tumor cells, with an acceptable toxicology profile. To gain more insight in the efficacy of TF-directed ADC treatment, we compared the internalization characteristics and intracellular routing of TF with the EGFR and HER2. Both in absence and presence of antibody, TF demonstrated more efficient internalization, lysosomal targeting, and degradation than EGFR and HER2. By conjugating TF, EGFR, and HER2-specific antibodies with duostatin-3, a toxin that induces potent cytotoxicity upon antibody-mediated internalization but lacks the ability to induce bystander killing, we were able to compare cytotoxicity of ADCs with different tumor specificities. TF-ADC demonstrated effective killing against tumor cell lines with variable levels of target expression. In xenograft models, TF-ADC was relatively potent in reducing tumor growth compared with EGFR- and HER2-ADCs. We hypothesize that the constant turnover of TF on tumor cells makes this protein specifically suitable for an ADC approach.


Subject(s)
Antineoplastic Agents/administration & dosage , ErbB Receptors/metabolism , Factor VIIa/metabolism , Immunotoxins/administration & dosage , Neoplasms, Experimental/drug therapy , Animals , Antibodies , Antineoplastic Agents/pharmacokinetics , Apoptosis , Cell Line, Tumor , Drug Delivery Systems , ErbB Receptors/immunology , Factor VIIa/immunology , Humans , Immunotoxins/pharmacokinetics , Lysosomes/metabolism , Mice , Neoplasms, Experimental/metabolism , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
15.
Anticancer Res ; 34(7): 3263-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24982330

ABSTRACT

BACKGROUND: We herein report a comparison of the radiolabels 177Lu and 125I bound to the monoclonal antibody HH1 that targets the CD37 antigen expressed on non-Hodgkin B-cell lymphomas. MATERIALS AND METHODS: Mixtures of 177Lu and 125I-labeled HH1 antibody were co-injected into nude mice carrying Ramos xenografts and the biodistribution using the paired label format allowing tracer comparisons in each individual mouse. RESULTS: Products of the two radionuclides had very similar immunoractivity in vitro but showed different properties in vivo. Both products had relevant stability in blood and most normal tissues in nude mice carrying subcutaneous Ramos xenografts. However, both the tumor uptake and retention were significantly higher for 177Lu vs. 125I labeled HH1. The tumor to normal tissue ratios were several-fold improved for 177Lu compared to radioiodine labeled antibodies. CONCLUSION: The data presented herein support the evaluation of CD37 as a target for clinical 177Lu-based radioimmunotherapy against b-cell malignancies.


Subject(s)
Immunotoxins/pharmacology , Iodine Radioisotopes/pharmacology , Lutetium/pharmacology , Lymphoma, B-Cell/radiotherapy , Radioisotopes/pharmacology , Radiopharmaceuticals/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Female , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacology , Humans , Immunotoxins/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Lutetium/pharmacokinetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Mice , Mice, Nude , Molecular Targeted Therapy , Radioimmunotherapy/methods , Radioisotopes/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Tetraspanins/immunology , Tissue Distribution , Xenograft Model Antitumor Assays
16.
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
17.
Cancer Biother Radiopharm ; 29(2): 83-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24383763

ABSTRACT

Time-activity uptake curves [u(t) in % injected dose per gram of tissue] may be described by different--often complicated--functional forms. Because of the need to readily compare sequences of engineered radiopharmaceuticals, it is efficient to use mean residence time (MRT) as a one-parameter descriptor. In applying this computation to a sequence of five cognate anti-carcinoembryonic antigen (CEA) antibodies, it was found that the intact form had the longest MRT in the blood with the other four cognates having values less by approximately a factor of 10 or more. This difference probably follows from the lack of an intact Fc segment on the latter engineered molecules. MRT values for a sequence of six scFv-Fc engineered fragments demonstrated that the double mutant had the shortest blood residence time--30-fold less compared with the wild type. Whereas it is not possible to directly apply the MRT to nonbolus (tumor or organ) curves, a residence time (τ) may be assigned using the uptake function. Using τ, it was found that the intact (natural) form of the anti-CEA cognate set had the longest time at the tumor site in the human xenograft model in nude mice. The MRT and τ concept are proposed to also allow comparison of possible relative blood and tissue exposures, respectively, for cognate sets of unlabeled engineered antibodies used to treat malignancies although no data are yet available in the literature for this application.


Subject(s)
Antibodies, Monoclonal/immunology , Carcinoembryonic Antigen/immunology , Immunotoxins/pharmacology , Immunotoxins/pharmacokinetics , Models, Biological , Radiopharmaceuticals/pharmacology , Radiopharmaceuticals/pharmacokinetics , Animals , Antibodies, Monoclonal/pharmacokinetics , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
18.
Radiother Oncol ; 110(2): 362-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24440046

ABSTRACT

PURPOSE: The effect of radioimmunotherapy (RIT) using the therapeutic radionuclide Y-90 bound to the anti-EGFR antibody cetuximab combined with external beam irradiation (EBRT) (EBRIT) on permanent local tumor control in vivo was examined. METHODS: Growth delay was evaluated in three human squamous cell carcinoma models after RIT with [(90)Y]Y-(CHX-A''-DTPA)4-cetuximab (Y-90-cetuximab). The EBRT dose required to cure 50% of the tumors (TCD50) for EBRT alone or EBRIT was evaluated in one RIT-responder (FaDu) and one RIT-non-responder (UT-SCC-5). EGFR expression and microenvironmental parameters were evaluated in untreated tumors, bioavailability was visualized by PET using ([(86)Y]Y-(CHX-A''-DTPA)4-cetuximab (Y-86-cetuximab) and biodistribution using Y-90-cetuximab. RESULTS: In UT-SCC-8 and FaDu but not in UT-SCC-5 radiolabeled cetuximab led to significant tumor growth delay. TCD50 after EBRT was significantly decreased by EGFR-targeted RIT in FaDu but not in UT-SCC-5. In contrast to EGFR expression, parameters of the tumor micromilieu and in particular the Y-90-cetuximab biodistribution or Y-86-cetuximab visualization in PET correlated with the responsiveness to RIT or EBRIT. CONCLUSION: EGFR-targeted EBRIT can improve permanent local tumor control compared to EBRT alone. PET imaging of bioavailability of labeled cetuximab appears to be a suitable predictor for response to EBRIT. This theragnostic approach should be further explored for clinical translation.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , ErbB Receptors/immunology , Immunotoxins/administration & dosage , Yttrium Radioisotopes/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Carcinoma, Squamous Cell/immunology , Cell Line, Tumor , Cetuximab , Female , Humans , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Isothiocyanates/chemistry , Male , Mice , Mice, Nude , Pentetic Acid/analogs & derivatives , Pentetic Acid/chemistry , Radioimmunotherapy , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/immunology , Random Allocation , Tissue Distribution , Yttrium Radioisotopes/chemistry , Yttrium Radioisotopes/pharmacokinetics
19.
J Cancer Res Clin Oncol ; 140(1): 35-43, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24132461

ABSTRACT

PURPOSE: The extradomain B of fibronectin (ED-B) is a promising vascular target for selective pharmacodelivery in cancer patients. We analyzed a large series of prostatectomies from patients with prostate cancer, hyperplastic prostate disease, and normal prostates to study extent and tumor-selectivity of ED-B expression. METHODS: Using immunohistology, 68 adenocarcinomas of the prostate or prostate cancer-inflicted lymph nodes, 4 samples of benign prostatic hyperplasia, and 6 normal prostate glands were studied for ED-B expressing newly formed blood vessels. Further, we treated an advanced prostate cancer patient with the anti-ED-B antibody (131)I-L19SIP to study in vivo target accessibility. RESULTS: ED-B-positive blood vessels were found significantly more frequent in prostate cancers as compared with peritumoral prostate tissues or normal prostate glands, independent of tumor differentiation. The ED-B-positive blood vessels' density was 97 (±23), 65 (±9), and 59 (±9)/mm(2) in G3, G2, and G1 prostate cancers, respectively, and 7 (±5)/mm(2) in normal prostate glands. In high-grade (G3) prostate cancers, also the peritumoral tissue showed a higher density of ED-B vessels than normal prostate glands. Similar results were obtained when ED-B-positive vessel density was expressed as a fraction of CD34-positive vessel density. Finally, selective uptake of ED-B-binding (131)I-L19SIP to tumor lesions was found in an advanced prostate cancer patient by whole-body planar scintigraphy. CONCLUSIONS: ED-B-positive blood vessels were found to a large extent in prostate cancer tissues, but only rarely in normal prostates or benign prostatic hyperplasia. Whole-body planar scintigraphy in a prostate cancer patient confirmed selective uptake of (131)I-L19SIP in the prostate cancer tissues, qualifying ED-B as a promising target for selective pharmacodelivery of anticancer agents in prostate cancer.


Subject(s)
Fibronectins/biosynthesis , Immunotoxins/pharmacokinetics , Iodine Radioisotopes/pharmacokinetics , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Antibodies/immunology , Blood Vessels/metabolism , Epitopes/analysis , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/metabolism , Paraffin Embedding , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Radioimmunodetection , Recombinant Fusion Proteins/immunology
20.
Cancer Biother Radiopharm ; 29(2): 70-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24299067

ABSTRACT

Carcinoembryonic antigen (CEA) is an attractive target molecule of radioimmunotherapy (RIT). To enhance RIT's therapeutic efficacy, the fractionation of radiolabeled antibody doses is an attractive strategy. In this study, a fully human anti-CEA monoclonal antibody (mAb) C2-45 was selected by virtue of its lack of immunogenicity, and the effectiveness of fractionated RIT with yttrium-90 (9°Y)-labeled mAb C2-45 was evaluated. In LS180 tumor-bearing mice, indium-111 (¹¹¹In)-labeled mAb C2-45 showed high and persistent tumor accumulation. Therapeutic studies were performed with single doses of 9°Y-mAb C2-45 (100 or 200 µCi) or double doses of 100 µCi 9°Y-mAb C2-45 at different intervals (5, 10, and 15 days). All 9°Y-mAb C2-45-treated mice showed inhibition of tumor progression, while the time to tumor progression was much longer in both the 200-µCi-treated group and the double 100-µCi-treated group than in the single 100-µCi-treated group. The therapeutic effect of the double 100 µCi administration at days 0 and 15 lasted significantly longer than that in the other treatment groups. These findings indicate that 9°Y-mAb C2-45 may be a promising agent for the treatment of CEA-positive cancer and that the fractionation of 9°Y-labeled antibody doses could enhance the therapeutic effect if performed according to an appropriate protocol.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoembryonic Antigen/immunology , Colonic Neoplasms/radiotherapy , Immunotoxins/administration & dosage , Radioimmunotherapy/methods , Radiopharmaceuticals/administration & dosage , Yttrium Radioisotopes/administration & dosage , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Humans , Immunotoxins/immunology , Immunotoxins/pharmacokinetics , Mice , Mice, Inbred BALB C , Mice, Nude , Radiopharmaceuticals/pharmacokinetics , Random Allocation , Tissue Distribution , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...