Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
2.
Cancers (Basel) ; 14(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36010941

ABSTRACT

Metastatic prostate cancer is a lethal disease in patients incapable of responding to therapeutic interventions. Invasive prostate cancer spread is caused by failure of the normal anti-cancer defense systems that are controlled by the tumour suppressor protein, p53. Upon mutation, p53 malfunctions. Therapeutic strategies to directly re-empower the growth-restrictive capacities of p53 in cancers have largely been unsuccessful, frequently because of a failure to discriminate responses in diseased and healthy tissues. Our studies sought alternative prostate cancer drivers, intending to uncover new treatment targets. We discovered the oncogenic potency of MDM4 in prostate cancer cells, both in the presence and absence of p53 and also its mutation. We uncovered that sustained depletion of MDM4 is growth inhibitory in prostate cancer cells, involving either apoptosis or senescence, depending on the cell and genetic context. We identified that the potency of MDM4 targeting could be potentiated in prostate cancers with mutant p53 through the addition of a first-in-class small molecule drug that was selected as a p53 reactivator and has the capacity to elevate oxidative stress in cancer cells to drive their death.

3.
Nat Commun ; 12(1): 7057, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34862374

ABSTRACT

In response to genotoxic stress, the tumor suppressor p53 acts as a transcription factor by regulating the expression of genes critical for cancer prevention. Mutations in the gene encoding p53 are associated with cancer development. PRIMA-1 and eprenetapopt (APR-246/PRIMA-1MET) are small molecules that are converted into the biologically active compound, methylene quinuclidinone (MQ), shown to reactivate mutant p53 by binding covalently to cysteine residues. Here, we investigate the structural basis of mutant p53 reactivation by MQ based on a series of high-resolution crystal structures of cancer-related and wild-type p53 core domains bound to MQ in their free state and in complexes with their DNA response elements. Our data demonstrate that MQ binds to several cysteine residues located at the surface of the core domain. The structures reveal a large diversity in MQ interaction modes that stabilize p53 and its complexes with DNA, leading to a common global effect that is pertinent to the restoration of non-functional p53 proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Aza Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Neoplasms/drug therapy , Quinuclidines/pharmacology , Tumor Suppressor Protein p53/agonists , Antineoplastic Agents/therapeutic use , Aza Compounds/chemistry , Aza Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Crystallography, X-Ray , Humans , Loss of Function Mutation/drug effects , Neoplasms/genetics , Protein Domains/drug effects , Quinuclidines/chemistry , Quinuclidines/therapeutic use , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/isolation & purification , Tumor Suppressor Protein p53/ultrastructure
4.
Cell Death Dis ; 12(7): 709, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267184

ABSTRACT

Asparaginase depletes extracellular asparagine in the blood and is an important treatment for acute lymphoblastic leukemia (ALL) due to asparagine auxotrophy of ALL blasts. Unfortunately, resistance occurs and has been linked to expression of the enzyme asparagine synthetase (ASNS), which generates asparagine from intracellular sources. Although TP53 is the most frequently mutated gene in cancer overall, TP53 mutations are rare in ALL. However, TP53 mutation is associated with poor therapy response and occurs at higher frequency in relapsed ALL. The mutant p53-reactivating compound APR-246 (Eprenetapopt/PRIMA-1Met) is currently being tested in phase II and III clinical trials in several hematological malignancies with mutant TP53. Here we present CEllular Thermal Shift Assay (CETSA) data indicating that ASNS is a direct or indirect target of APR-246 via the active product methylene quinuclidinone (MQ). Furthermore, combination treatment with asparaginase and APR-246 resulted in synergistic growth suppression in ALL cell lines. Our results thus suggest a potential novel treatment strategy for ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Asparaginase/pharmacology , Cell Proliferation/drug effects , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Quinuclidines/pharmacology , Tumor Suppressor Protein p53/agonists , Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
EMBO Mol Med ; 13(2): e10852, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33314700

ABSTRACT

The tumor suppressor gene TP53 is the most frequently mutated gene in cancer. The compound APR-246 (PRIMA-1Met/Eprenetapopt) is converted to methylene quinuclidinone (MQ) that targets mutant p53 protein and perturbs cellular antioxidant balance. APR-246 is currently tested in a phase III clinical trial in myelodysplastic syndrome (MDS). By in vitro, ex vivo, and in vivo models, we show that combined treatment with APR-246 and inhibitors of efflux pump MRP1/ABCC1 results in synergistic tumor cell death, which is more pronounced in TP53 mutant cells. This is associated with altered cellular thiol status and increased intracellular glutathione-conjugated MQ (GS-MQ). Due to the reversibility of MQ conjugation, GS-MQ forms an intracellular drug reservoir that increases availability of MQ for targeting mutant p53. Our study shows that redox homeostasis is a critical determinant of the response to mutant p53-targeted cancer therapy.


Subject(s)
Neoplasms , Pharmaceutical Preparations , Cell Death , Cell Line, Tumor , Humans , Mutation , Neoplasms/drug therapy , Quinuclidines , Sulfhydryl Compounds , Tumor Suppressor Protein p53/genetics
6.
Nat Commun ; 8: 14844, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28348409

ABSTRACT

TP53, a critical tumour suppressor gene, is mutated in over half of all cancers resulting in mutant-p53 protein accumulation and poor patient survival. Therapeutic strategies to target mutant-p53 cancers are urgently needed. We show that accumulated mutant-p53 protein suppresses the expression of SLC7A11, a component of the cystine/glutamate antiporter, system xC-, through binding to the master antioxidant transcription factor NRF2. This diminishes glutathione synthesis, rendering mutant-p53 tumours susceptible to oxidative damage. System xC- inhibitors specifically exploit this vulnerability to preferentially kill cancer cells with stabilized mutant-p53 protein. Moreover, we demonstrate that SLC7A11 expression is a novel and robust predictive biomarker for APR-246, a first-in-class mutant-p53 reactivator that also binds and depletes glutathione in tumours, triggering lipid peroxidative cell death. Importantly, system xC- antagonism strongly synergizes with APR-246 to induce apoptosis in mutant-p53 tumours. We propose a new paradigm for targeting cancers that accumulate mutant-p53 protein by inhibiting the SLC7A11-glutathione axis.


Subject(s)
Amino Acid Transport System y+/metabolism , Glutathione/metabolism , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/genetics , Apoptosis/drug effects , Cell Line, Tumor , Humans , Lipid Peroxidation/drug effects , Models, Biological , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Quinuclidines/pharmacology , Reactive Oxygen Species/metabolism , Stress, Physiological/drug effects
7.
Front Oncol ; 6: 21, 2016.
Article in English | MEDLINE | ID: mdl-26870698

ABSTRACT

TP53 is the most frequently mutated gene in cancer. The p53 protein activates transcription of genes that promote cell cycle arrest or apoptosis, or regulate cell metabolism, and other processes. Missense mutations in TP53 abolish specific DNA binding of p53 and allow evasion of apoptosis and accelerated tumor progression. Mutant p53 often accumulates at high levels in tumor cells. Pharmacological reactivation of mutant p53 has emerged as a promising strategy for improved cancer therapy. Small molecules that restore wild type activity of mutant p53 have been identified using various approaches. One of these molecules, APR-246, is a prodrug that is converted to the Michael acceptor methylene quinuclidinone (MQ) that binds covalently to cysteines in p53, leading to refolding and restoration of wild type p53 function. MQ also targets the cellular redox balance by inhibiting thioredoxin reductase (TrxR1) and depleting glutathione. This dual mechanism of action may account for the striking synergy between APR-246 and platinum compounds. APR-246 is the only mutant p53-targeting compound in clinical development. A phase I/IIa clinical trial in hematological malignancies and prostate cancer showed good safety profile and clinical effects in some patients. APR-246 is currently tested in a phase Ib/II trial in patients with high-grade serous ovarian cancer.

8.
Acta Oncol ; 55(2): 140-8, 2016.
Article in English | MEDLINE | ID: mdl-26161618

ABSTRACT

BACKGROUND: A phase Ia/b dose-escalation study was performed to characterize the safety, efficacy and pharmacokinetic properties of the oral small molecule insulin-like growth factor-1-receptor pathway modulator AXL1717 in patients with advanced solid tumors. MATERIAL AND METHODS: This was a prospective, single-armed, open label, dose-finding phase Ia/b study with the aim of single day dosing (phase Ia) to define the starting dose for multi-day dosing (phase Ib), and phase Ib to define and confirm recommended phase II dose (RP2D) and if possible maximum tolerated dose (MTD) for repeated dosing. RESULTS AND CONCLUSION: Phase Ia enrolled 16 patients and dose escalations up to 2900 mg BID were successfully performed without any dose limiting toxicity (DLT). A total of 39 patients were treated in phase Ib. AXL1717 was well tolerated with neutropenia as the only dose-related, reversible, DLT. RP2D dose was found to be 390 mg BID for four weeks. Some patients, mainly with NSCLC, demonstrated signs of clinical benefit, including four partial tumor responses (one according to RECIST and three according to PET). The 15 patients with NSCLC with treatment duration longer than two weeks with single agent AXL1717 in third or fourth line of therapy showed a median progression-free survival of 31 weeks and overall survival of 60 weeks. Down-regulation of IGF-1R on granulocytes and increases of free serum levels of IGF-1 were seen in patients treated with AXL1717. AXL1717 had an acceptable safety profile and demonstrated promising efficacy in this heavily pretreated patient cohort, especially in patients with NSCLC. RP2D was concluded to be 390 mg BID for four weeks. Trial number is NCT01062620.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Podophyllotoxin/analogs & derivatives , Receptor, IGF Type 1/metabolism , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Disease-Free Survival , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Maximum Tolerated Dose , Middle Aged , Molecular Targeted Therapy , Neutropenia/chemically induced , Podophyllotoxin/administration & dosage , Podophyllotoxin/adverse effects , Podophyllotoxin/blood , Podophyllotoxin/therapeutic use , Prospective Studies , Signal Transduction/drug effects , Treatment Outcome
9.
Biomed Res Int ; 2015: 605427, 2015.
Article in English | MEDLINE | ID: mdl-26075248

ABSTRACT

In drug development, the "onus" of the low R&D efficiency has been put traditionally onto the drug discovery process (i.e., finding the right target or "binding" functionality). Here, we show that manufacturing is not only a central component of product success, but also that, by integrating manufacturing and discovery activities in a "holistic" interpretation of QbD methodologies, we could expect to increase the efficiency of the drug discovery process as a whole. In this new context, early risk assessment, using developability methodologies and computational methods in particular, can assist in reducing risks during development in a cost-effective way. We define specific areas of risk and how they can impact product quality in a broad sense, including essential aspects such as product efficacy and patient safety. Emerging industry practices around developability are introduced, including some specific examples of applications to biotherapeutics. Furthermore, we suggest some potential workflows to illustrate how developability strategies can be introduced in practical terms during early drug development in order to mitigate risks, reduce drug attrition and ultimately increase the robustness of the biopharmaceutical supply chain. Finally, we also discuss how the implementation of such methodologies could accelerate the access of new therapeutic treatments to patients in the clinic.


Subject(s)
Biopharmaceutics/methods , Drug Design , Drug Industry/methods , Humans
10.
Proc Natl Acad Sci U S A ; 111(48): 17110-5, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25406323

ABSTRACT

Proteins endocytosed from serum are degraded in the lysosomes. However, serum albumin (SA) and IgG, through its Fc part, bind to the neonatal Fc receptor (FcRn) at low pH in the endosome after endocytosis, and are transported back to the cellular surface, where they are released into the bloodstream, resulting in an extended serum circulation time. Association with Fc or SA has been used to prolong the in vivo half-life of biopharmaceuticals, using the interaction with FcRn to improve treatment regimens. This has been achieved either directly, by fusion or conjugation to Fc or SA, or indirectly, using SA-binding proteins. The present work takes this principle one step further, presenting small affinity proteins that bind directly to FcRn, mediating extension of the serum half-life of fused biomolecules. Phage display technology was used to select affibody molecules that can bind to FcRn in the pH-dependent manner required for rescue by FcRn. The biophysical and binding properties were characterized in vitro, and the affibody molecules were found to bind to FcRn more strongly at low pH than at neutral pH. Attachment of the affibody molecules to a recombinant protein, already engineered for increased half-life, resulted in a nearly threefold longer half-life in mice. These tags should have general use as fusion partners to biopharmaceuticals to extend their half-lives in vivo.


Subject(s)
Carrier Proteins/metabolism , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Animals , Binding, Competitive , Carrier Proteins/genetics , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Half-Life , HeLa Cells , Histocompatibility Antigens Class I/genetics , Humans , Hydrogen-Ion Concentration , Male , Mice, Inbred Strains , Peptide Library , Protein Binding , Receptors, Fc/genetics , Recombinant Fusion Proteins/blood
11.
Oncotarget ; 5(18): 8379-92, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25268741

ABSTRACT

Picropodophyllin (PPP) is an anticancer drug undergoing clinical development in NSCLC. PPP has been shown to suppress IGF-1R signaling and to induce a G2/M cell cycle phase arrest but the exact mechanisms remain to be elucidated. The present study identified an IGF-1-independent mechanism of PPP leading to pro-metaphase arrest. The mitotic block was induced in human cancer cell lines and in an A549 xenograft mouse but did not occur in normal hepatocytes/mouse tissues. Cell cycle arrest by PPP occurred in vitro and in vivo accompanied by prominent CDK1 activation, and was IGF-1R-independent since it occurred also in IGF-1R-depleted and null cells. The tumor cells were not arrested in G2/M but in mitosis. Centrosome separation was prevented during mitotic entry, resulting in a monopolar mitotic spindle with subsequent prometaphase-arrest, independent of Plk1/Aurora A or Eg5, and leading to cell features of mitotic catastrophe. PPP also increased soluble tubulin and decreased spindle-associated tubulin within minutes, indicating that it interfered with microtubule dynamics. These results provide a novel IGF-1R-independent mechanism of antitumor effects of PPP.


Subject(s)
Antineoplastic Agents/pharmacology , Centrosome/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Lung Neoplasms/drug therapy , Microtubules/drug effects , Mitosis/drug effects , Podophyllotoxin/analogs & derivatives , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , CDC2 Protein Kinase , Cell Survival/drug effects , Centrosome/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Enzyme Activation , Hep G2 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MCF-7 Cells , Microtubules/metabolism , Podophyllotoxin/pharmacology , RNA Interference , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Time Factors , Transfection , Tubulin/metabolism , Xenograft Model Antitumor Assays
12.
Mol Pharm ; 11(11): 3957-64, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-24972112

ABSTRACT

Platelet-derived growth factor receptor ß (PDGFRß) is a transmembrane tyrosine kinase receptor involved, for example, in angiogenesis. Overexpression and excessive signaling of PDGFRß has been observed in multiple malignant tumors and fibrotic diseases, making this receptor a pharmaceutical target for monoclonal antibodies and tyrosine kinase inhibitors. Successful targeted therapy requires identification of responding patients. Radionuclide molecular imaging would enable determination of the PDGFRß status in all lesions using a single noninvasive repeatable procedure. Recently, we have demonstrated that the affibody molecule Z09591 labeled with (111)In can specifically target PDGFRß-expressing tumors in vivo. The use of positron emission tomography (PET) as an imaging technique would provide superior resolution, sensitivity, and quantitation accuracy. In this study, a DOTA-conjugated Z09591 was labeled with the generator-produced positron emitting radionuclide (68)Ga (T1/2 = 67.6 min, Eß + max = 1899 keV, 89% ß(+)). (68)Ga-DOTA-Z09591 retained the capacity to specifically bind to PDGFRß-expressing U-87 MG glioma cells. The half-maximum inhibition concentration (IC50) of (68)Ga-DOTA-Z09591 (6.6 ± 1.4 nM) was somewhat higher than that of (111)In-DOTA-Z09591 (1.4 ± 1.2 nM). (68)Ga-DOTA-Z09591 demonstrated specific (saturable) targeting of U-87 MG xenografts in immunodeficient mice. The tumor uptake at 2 h after injection was 3.7 ± 1.7% IA/g, which provided a tumor-to-blood ratio of 8.0 ± 3.1. The only organ with higher accumulation of radioactivity was the kidney. MicroPET imaging provided high-contrast imaging of U-87 MG xenografts. In conclusion, the (68)Ga-labeled affibody molecule Z09591 is a promising candidate for further development as a probe for imaging PDGFRß expression in vivo using PET.


Subject(s)
Gallium Radioisotopes , Positron-Emission Tomography , Receptor, Platelet-Derived Growth Factor beta/chemistry , Animals , Antibodies, Monoclonal/chemistry , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Female , Gallium Radioisotopes/chemistry , Glioma/diagnostic imaging , Heterocyclic Compounds, 1-Ring/chemistry , Indium Radioisotopes , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Multimodal Imaging , Neoplasm Transplantation , Neovascularization, Pathologic , Tomography, X-Ray Computed
13.
Nucl Med Commun ; 35(7): 767-76, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24709981

ABSTRACT

BACKGROUND: The number of radiolabeled monoclonal antibodies (mAbs) used for medical imaging and cancer therapy is increasing. The required chemical modification for attaching a radioactive label and all associated treatment may lead to a damaged mAb subpopulation. This paper describes a novel method, concentration through kinetics (CTK), for rapid assessment of the concentration of immunoreactive mAb and the specific radioactivity, based on monitoring binding kinetics. METHODS: The interaction of radiolabeled mAb with either the antigen or a general mAb binder such as Protein A was monitored in real time using the instrument LigandTracer. As the curvature of the binding trace has a distinct shape based on the interaction kinetics and concentration of the functional mAb, the immunoreactive mAb concentration could be calculated through reverse kinetic fitting of the binding curves, using software developed for this project. The specific activity, describing the degree of radioactive labeling, was determined through the use of calibrated signal intensities. RESULTS: The performance of the CTK assay was evaluated on the basis of various mAb-based interaction systems and assay formats, and it was shown that the assay can provide accurate and repeatable results for immunoreactive concentration and specific activity, with both accuracy and relative SD values below 15%. CONCLUSION: By applying reverse kinetics on real-time binding traces it is possible to estimate the functional concentration and specific activity of radiolabeled mAb. The CTK assay may in the future be included as a complement to current quality assessment methods of radiolabeled mAbs.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Immunoconjugates/chemistry , Immunoconjugates/immunology , Radiochemistry/methods , Adsorption , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antigens, Neoplasm/immunology , Automation , Dextrans/chemistry , Kinetics , Plastics/chemistry
14.
Biopolymers ; 102(3): 252-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24549714

ABSTRACT

Glucagon-like peptide 1 (GLP-1) and related peptide agonists have been extensively investigated for glycaemic control in Type 2 diabetes, and may also have therapeutic applications for other diseases. Due to the short half-life (t1/2 < 2 min) of the endogenous peptide, caused by proteolytic degradation and renal clearance, different strategies for half-life extension and sustained release have been explored. In the present study, conjugates between a GLP-1 analogue and a 5 kDa albumin-binding domain (ABD) derived from streptococcal protein G have been chemically synthesized and evaluated. ABD binds with high affinity to human serum albumin, which is highly abundant in plasma and functions as a drug carrier in the circulation. Three different GLP-1-ABD conjugates, with the two peptides connected by linkers of two, four, and six PEG units, respectively, were synthesized and tested in mouse pancreatic islets at high (11 mM) and low (3 mM) glucose concentration. Insulin release upon stimulation was shown to be glucose-dependent, showing no significant difference between the three different GLP-1-ABD conjugates and unconjugated GLP-1 analogue. The biological activity, in combination with the high affinity binding to albumin, make the GLP-1-ABD conjugates promising GLP-1 receptor agonists expected to show extended in vivo half-life.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/chemistry , Serum Albumin/chemistry , Animals , Biosensing Techniques , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Half-Life , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans , Mice, Obese , Peptides/chemical synthesis , Peptides/chemistry , Protein Structure, Tertiary , Rats
15.
J Nucl Med ; 55(2): 294-300, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24408895

ABSTRACT

UNLABELLED: The overexpression and excessive signaling of platelet-derived growth factor receptor ß (PDGFRß) has been detected in cancers, atherosclerosis, and a variety of fibrotic diseases. Radionuclide in vivo visualization of PDGFRß expression might help to select PDGFRß targeting treatment for these diseases. The goal of this study was to evaluate the feasibility of in vivo radionuclide imaging of PDGFRß expression using an Affibody molecule, a small nonimmunoglobulin affinity protein. METHODS: The PDGFRß-binding Z09591 Affibody molecule was site-specifically conjugated with a maleimido derivative of DOTA and labeled with (111)In. Targeting of the PDGFRß-expressing U-87 MG glioblastoma cell line using (111)In-DOTA-Z09591 was evaluated in vitro and in vivo. RESULTS: DOTA-Z09591 was stably labeled with (111)In with preserved specific binding to PDGFRß-expressing cells in vitro. The dissociation constant for (111)In-DOTA-Z09591 binding to U-87 MG cells was determined to be 92 ± 10 pM. In mice bearing U-87 MG xenografts, the tumor uptake of (111)In-DOTA-Z09591 was 7.2 ± 2.4 percentage injected dose per gram and the tumor-to-blood ratio was 28 ± 14 at 2 h after injection. In vivo receptor saturation experiments demonstrated that targeting of U-87 MG xenografts in mice was PDGFRß-specific. U-87 MG xenografts were clearly visualized using small-animal SPECT/CT at 3 h after injection. CONCLUSION: This study demonstrates the feasibility of in vivo visualization of PDGFRß-expressing xenografts using an Affibody molecule. Further development of radiolabeled Affibody molecules might provide a useful clinical imaging tool for PDGFRß expression during various pathologic conditions.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Coordination Complexes/chemistry , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Heterocyclic Compounds, 1-Ring/metabolism , Indium Radioisotopes/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Fusion Proteins/chemistry , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Multimodal Imaging/methods , Neoplasm Transplantation , Positron-Emission Tomography/methods , Protein Binding , Protein Structure, Tertiary , Tomography, Emission-Computed, Single-Photon/methods , Tomography, X-Ray Computed/methods
16.
Protein Eng Des Sel ; 26(10): 635-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23924760

ABSTRACT

Affibody molecules are engineered binding proteins, in which the three-helix bundle motif of the Z domain derived from protein A is used as a scaffold for sequence variation. We used phage display to select Affibody binders to staphylococcal protein A itself. The best binder, called ZpA963, binds with similar affinity and kinetics to the five homologous E, D, A, B and C domains of protein A, and to a five-domain protein A construct with an average dissociation constant, K(D), of ~20 nM. The structure of ZpA963 in complex with the Z domain shows that it interacts with a surface on Z that is identical in the five protein A domains, which explains the multi-domain affinity. This property allows for high-affinity binding by dimeric Affibody molecules that simultaneously engage two protein A domains in a complex. We studied two ZpA963 dimers in which the subunits were linked by a C-terminal disulfide in a symmetric dimer or head-to-tail in a fusion protein, respectively. The dimers both bind protein A with high affinity, very slow off-rates and with saturation-dependent kinetics that can be understood in terms of dimer binding to multiple sites. The head-to-tail (ZpA963)2htt dimer binds with an off-rate of k(off) ≤ 5 × 10(-6) s(-1) and an estimated K(D) ≤ 16 pM. The results illustrate how dimers of selected monomer binding proteins can provide an efficient route for engineering of high-affinity binders to targets that contain multiple homologous domains or repeated structural units.


Subject(s)
Protein Engineering , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Staphylococcal Protein A/metabolism , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Peptide Library , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics
17.
J Nucl Med ; 54(6): 961-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23528382

ABSTRACT

UNLABELLED: Because of their better penetration, smaller targeting proteins may be superior to antibodies for radioimmunotherapy of solid tumors. Therefore, Affibody molecules (6.5 kDa) have a potential for being suitable as targeted moiety for radiolabeled therapeutic proteins. Previous studies have demonstrated that a fusion of an Affibody molecule with an albumin-binding domain (ABD) provides a strong noncovalent binding to albumin in vivo. This strong noncovalent binding can be used for reduction of the renal uptake of the Affibody molecule while maintaining a size smaller than that of an antibody, which is important when using residualizing radionuclide labels conjugated to Affibody molecules. The goal of this study was to design and evaluate a new targeting Affibody-ABD fusion protein with improved biodistribution properties for radionuclide therapy. METHODS: A novel Affibody-based construct, ZHER2:2891-ABD035-DOTA (ABY-027), was created by fusion of the reengineered HER2-binding Affibody molecule ZHER2:2891 to the N terminus of the high-affinity ABD035, and a maleimido-derivative of DOTA was conjugated at the C terminus of the construct. Binding and processing of (177)Lu-ABY-027 by HER2-expressing cells were evaluated in vitro. Targeting of HER2-expressing SKOV-3 xenografts was evaluated in BALB/C nu/nu mice and compared with targeting of previously reported ABD-(ZHER2:342)2. RESULTS: The binding affinity (dissociation constant) of ABY-027 to HER2 (74 pM) was the same as for the parental ZHER2:2891 (76 pM). ABY-027 was stably labeled with (177)Lu and (111)In with preserved specific binding to HER2-expressing cells in vitro. In vivo receptor saturation experiments demonstrated that targeting of SKOV-3 xenografts in BALB/C nu/nu mice was HER2-specific. (177)Lu-ABY-027 demonstrated substantially (2- to 3-fold) lower renal and hepatic uptake than previously assessed HER2-specific Affibody-based albumin-binding agents. Tumor uptake of radiolabeled ABY-027 at 48 h after injection was 2-fold higher than that for previously reported ABD-(ZHER2:342)2. CONCLUSION: An optimized molecular design of an ABD fusion protein resulted in an Affibody molecule construct with better properties for therapy. Fully preserved in vivo targeting of the fusion protein was shown in xenografted mice. Site-specific coupling of DOTA provides a uniform conjugate and creates the potential for labeling with a broad range of therapeutic radionuclides. The biodistribution of (177)Lu-ABY-027 in a murine model suggests it is more suitable for therapy than alternative approaches.


Subject(s)
Albumins/metabolism , Lutetium/therapeutic use , Radioisotopes/therapeutic use , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Animals , Binding Sites , Cell Line, Tumor , Female , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Isotope Labeling , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/radiotherapy , Protein Structure, Tertiary , Protein Transport , Radiometry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Substrate Specificity , Tissue Distribution
18.
J Nucl Med ; 53(6): 953-60, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22586147

ABSTRACT

UNLABELLED: Radionuclide imaging of cancer-associated molecular alterations may contribute to patient stratification for targeting therapy. Scaffold high-affinity proteins, such as Affibody molecules, are a new, promising class of probes for in vivo imaging. METHODS: The effects of human epidermal growth factor receptor 2 (HER2) affinity and binding site composition of HER2-binding Affibody molecules, and of the HER2 density on the tumor targeting, were studied in vivo. The tumor uptake and tumor-to-organ ratios of Affibody molecules with moderate (dissociation constant [K(D)] = 10(-9) M) or high (K(D) = 10(-10) M) affinity were compared between tumor xenografts with a high (SKOV-3) and low (LS174T) HER2 expression level in BALB/C nu/nu mice. Two Affibody molecules with similar affinity (K(D) = 10(-10) M) but having alternative amino acids in the binding site were compared. RESULTS: In SKOV-3 xenografts, uptake was independent of affinity at 4 h after injection, but high-affinity binders provided 2-fold-higher tumor radioactivity retention at 24 h. In LS174T xenografts, uptake of high-affinity probes was already severalfold higher at 4 h after injection, and the difference was increased at 24 h. The clearance rate and tumor-to-organ ratios were influenced by the amino acid composition of the binding surface of the tracer protein. CONCLUSION: The optimal affinity of HER2-binding Affibody molecules depends on the expression of a molecular target. At a high expression level (>10(6) receptors per cell), an affinity in the low-nanomolar range is sufficient. At moderate expression, subnanomolar affinity is desirable. The binding site composition can influence the imaging contrast. This information may be useful for development of imaging agents based on scaffold affinity proteins.


Subject(s)
Indium Radioisotopes , Neoplasms, Experimental/diagnostic imaging , Receptor, ErbB-2/metabolism , Amino Acid Sequence , Animals , Binding Sites , Female , Isotope Labeling , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Radionuclide Imaging
19.
Chembiochem ; 13(7): 1024-31, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22532524

ABSTRACT

Affinity molecules labeled with different reporter groups, such as fluorophores or radionuclides, are valuable research tools used in a variety of applications. One class of engineered affinity proteins is Affibody molecules, which are small (6.5 kDa) proteins that can be produced by solid phase peptide synthesis (SPPS), thereby allowing site-specific incorporation of reporter groups during synthesis. The Affibody molecules are triple-helix proteins composed of a variable part, which gives the protein its binding specificity, and a constant part, which is identical for all Affibody molecules. In the present study, native chemical ligation (NCL) has been applied for combinatorial assembly of Affibody molecules from peptide fragments produced by Fmoc SPPS. The concept is demonstrated for the synthesis of three different Affibody molecules. The cysteine residue introduced at the site of ligation can be used for directed immobilization and does not interfere with the function of the investigated proteins. This strategy combines a high-yield production method with facilitated preparation of proteins with different C-terminal modifications.


Subject(s)
Antibodies/chemistry , Biomimetic Materials/chemistry , Peptide Fragments/chemistry , Amino Acid Sequence , Antibodies/metabolism , Biomimetic Materials/chemical synthesis , Biomimetic Materials/metabolism , Combinatorial Chemistry Techniques , Humans , Immobilized Proteins/chemical synthesis , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Ligands , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Protein Engineering
20.
J Nucl Med ; 53(1): 90-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22173843

ABSTRACT

UNLABELLED: One of the pathways leading to androgen independence in prostate cancer involves upregulation of insulinlike growth factor type 1 receptor (IGF-1R). Radionuclide imaging of IGF-1R in tumors might be used for selection of patients who would most likely benefit from IGF-1R-targeted therapy. The goal of this study was to evaluate the feasibility of in vivo radionuclide imaging of IGF-1R expression in prostate cancer xenografts using a small nonimmunoglobulin-derived binding protein called an Affibody molecule. METHODS: The IGF-1R-binding Z(IGF1R:4551) Affibody molecule was site-specifically conjugated with a maleimido derivative of DOTA and labeled with (111)In. The binding of radiolabeled Z(IGF1R:4551) to IGF-1R-expressing cells was evaluated in vitro and in vivo. RESULTS: DOTA-Z(IGF1R:4551) can be stably labeled with (111)In with preserved specific binding to IGF-1R-expressing cells in vitro. In mice, (111)In-DOTA-Z(IGF1R:4551) accumulated in IGF-1R-expressing organs (pancreas, stomach, lung, and salivary gland). Receptor saturation experiments demonstrated that targeting of DU-145 prostate cancer xenografts in NMRI nu/nu mice was IGF-1R-specific. The tumor uptake was 1.1 ± 0.3 percentage injected dose per gram, and the tumor-to-blood ratio was 3.2 ± 0.2 at 8 h after injection. CONCLUSION: This study demonstrates the feasibility of in vivo targeting of IGF-1R-expressing prostate cancer xenografts using an Affibody molecule. Further development of radiolabeled Affibody molecules might provide a useful clinical tool for stratification of patients with prostate cancer for IGF-1R-targeting therapy.


Subject(s)
Cell Transformation, Neoplastic , Heterocyclic Compounds, 1-Ring/chemistry , Indium Radioisotopes , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Receptor, IGF Type 1/metabolism , Recombinant Fusion Proteins , Animals , Cell Line, Tumor , Feasibility Studies , Humans , Male , Mice , Prostatic Neoplasms/metabolism , Radionuclide Imaging , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...