Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
2.
ACS Pharmacol Transl Sci ; 5(11): 1156-1168, 2022 Nov 11.
Article de Anglais | MEDLINE | ID: mdl-36407952

RÉSUMÉ

Bruton's tyrosine kinase (BTK) is a member of the TEC-family kinases and crucial for the proliferation and differentiation of B-cells. We evaluated the therapeutic potential of a covalent inhibitor (JS25) with nanomolar potency against BTK and with a more desirable selectivity and inhibitory profile compared to the FDA-approved BTK inhibitors ibrutinib and acalabrutinib. Structural prediction of the BTK/JS25 complex revealed sequestration of Tyr551 that leads to BTK's inactivation. JS25 also inhibited the proliferation of myeloid and lymphoid B-cell cancer cell lines. Its therapeutic potential was further tested against ibrutinib in preclinical models of B-cell cancers. JS25 treatment induced a more pronounced cell death in a murine xenograft model of Burkitt's lymphoma, causing a 30-40% reduction of the subcutaneous tumor and an overall reduction in the percentage of metastasis and secondary tumor formation. In a patient model of diffuse large B-cell lymphoma, the drug response of JS25 was higher than that of ibrutinib, leading to a 64% "on-target" efficacy. Finally, in zebrafish patient-derived xenografts of chronic lymphocytic leukemia, JS25 was faster and more effective in decreasing tumor burden, producing superior therapeutic effects compared to ibrutinib. We expect JS25 to become therapeutically relevant as a BTK inhibitor and to find applications in the treatment of hematological cancers and other pathologies with unmet clinical treatment.

3.
Cancer Discov ; 12(2): 372-387, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34635570

RÉSUMÉ

Personalized medicine aims to match the right drug with the right patient by using specific features of the individual patient's tumor. However, current strategies of personalized therapy matching provide treatment opportunities for less than 10% of patients with cancer. A promising method may be drug profiling of patient biopsy specimens with single-cell resolution to directly quantify drug effects. We prospectively tested an image-based single-cell functional precision medicine (scFPM) approach to guide treatments in 143 patients with advanced aggressive hematologic cancers. Fifty-six patients (39%) were treated according to scFPM results. At a median follow-up of 23.9 months, 30 patients (54%) demonstrated a clinical benefit of more than 1.3-fold enhanced progression-free survival compared with their previous therapy. Twelve patients (40% of responders) experienced exceptional responses lasting three times longer than expected for their respective disease. We conclude that therapy matching by scFPM is clinically feasible and effective in advanced aggressive hematologic cancers. SIGNIFICANCE: This is the first precision medicine trial using a functional assay to instruct n-of-one therapies in oncology. It illustrates that for patients lacking standard therapies, high-content assay-based scFPM can have a significant value in clinical therapy guidance based on functional dependencies of each patient's cancer.See related commentary by Letai, p. 290.This article is highlighted in the In This Issue feature, p. 275.


Sujet(s)
Tumeurs hématologiques/traitement médicamenteux , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Autriche , Études de cohortes , Femelle , Tumeurs hématologiques/mortalité , Humains , Mâle , Adulte d'âge moyen , Thérapie moléculaire ciblée , Médecine de précision , Survie sans progression , Jeune adulte
4.
J Nucl Med ; 62(3): 360-365, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-32680925

RÉSUMÉ

99mTc-PHC-102 is a 99mTc-labeled derivative of acetazolamide, a high-affinity small organic ligand of carbonic anhydrase IX (CAIX). 99mTc-PHC-102 has previously shown favorable in vivo biodistribution properties in mouse models of CAIX-positive clear cell renal cell carcinoma (ccRCC) and colorectal cancer. In this study, we aimed to explore the targeting performance of 99mTc-PHC-102 in SPECT in patients with renal cell carcinoma while also assessing the safety and tolerability of the radiotracer. Methods: We studied 5 patients with localized or metastatic ccRCC in a microdosing regimen, after the administration of a 50-µg total of CAIX ligand and 600-800 MBq of 99mTc-PHC-102. Tissue distribution and residence time in normal organs and tumors were analyzed by serial SPECT/CT scans at 3 time points (30 min, 2 h, and 6 h) after intravenous administration. Results: In the 5 patients studied, 99mTc-PHC-102 was well tolerated and no study drug-related adverse events were recorded. In the stomach, kidneys, and gallbladder, the radiotracer showed a rapid initial uptake, which cleared over time. Localization of the study drug in primary tumors of 5 patients was observed, with favorable tumor-to-background ratios. 99mTc-PHC-102 SPECT/CT allowed the identification of 4 previously unknown lung and lymph node metastases in 2 patients. Conclusion:99mTc-PHC-102 is a promising SPECT tracer for the imaging of patients with ccRCC. This tracer has the potential to identify primary and metastatic lesions in different anatomic locations. 99mTc-PHC-102 might also serve as a companion diagnostic agent for future CAIX-targeting therapeutics.


Sujet(s)
Néphrocarcinome/imagerie diagnostique , Néphrocarcinome/anatomopathologie , Tumeurs du rein/imagerie diagnostique , Tumeurs du rein/anatomopathologie , Dose de rayonnement , Tomographie par émission monophotonique couplée à la tomodensitométrie , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Humains , Mâle , Adulte d'âge moyen , Métastase tumorale , Sécurité
5.
Curr Opin Chem Biol ; 56: 72-78, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32086157

RÉSUMÉ

Anticancer drug discovery and development using conventional cell line and animal models has traditionally had a low overall success rate. Despite yielding game-changing new therapeutics, 10-20 new molecules have to be brought to the clinic to obtain one new approval, making this approach costly and inefficient. The use of in vitro experimental models based on primary human tumour tissues has the potential to provide a representation of human cancer biology that is closer to an actual patient and to 'bridge the translational gap' between preclinical and clinical research. Here, we review recent advances in the use of human tumour samples for preclinical research through organoid development or as primary patient materials. While challenges still remain regarding analysis, validation and scalability, evidence is mounting for the applicability of both models as preclinical research tools.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs/imagerie diagnostique , Tumeurs/thérapie , Animaux , Évaluation préclinique de médicament , Tests de criblage d'agents antitumoraux , Tests de criblage à haut débit , Humains , Apprentissage machine , Modèles biologiques , Imagerie optique , Organoïdes/métabolisme , Analyse sur cellule unique
6.
Nat Chem Biol ; 15(3): 232-240, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30692684

RÉSUMÉ

The Bruton tyrosine kinase (BTK) inhibitor ibrutinib has substantially improved therapeutic options for chronic lymphocytic leukemia (CLL). Although ibrutinib is not curative, it has a profound effect on CLL cells and may create new pharmacologically exploitable vulnerabilities. To identify such vulnerabilities, we developed a systematic approach that combines epigenome profiling (charting the gene-regulatory basis of cell state) with single-cell chemosensitivity profiling (quantifying cell-type-specific drug response) and bioinformatic data integration. By applying our method to a cohort of matched patient samples collected before and during ibrutinib therapy, we identified characteristic ibrutinib-induced changes that provide a starting point for the rational design of ibrutinib combination therapies. Specifically, we observed and validated preferential sensitivity to proteasome, PLK1, and mTOR inhibitors during ibrutinib treatment. More generally, our study establishes a broadly applicable method for investigating treatment-specific vulnerabilities by integrating the complementary perspectives of epigenetic cell states and phenotypic drug responses in primary patient samples.


Sujet(s)
Agammaglobulinaemia tyrosine kinase/métabolisme , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Adénine/analogues et dérivés , Agammaglobulinaemia tyrosine kinase/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Chromatine/physiologie , Association médicamenteuse , Résistance aux médicaments antinéoplasiques/génétique , Épigenèse génétique/génétique , Épigénomique/méthodes , Humains , Leucémie chronique lymphocytaire à cellules B/génétique , Pipéridines , Inhibiteurs de protéines kinases , Protein-Serine-Threonine Kinases/métabolisme , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/physiologie , Protéines proto-oncogènes/métabolisme , Pyrazoles/pharmacologie , Pyrimidines/pharmacologie , Transduction du signal , Analyse sur cellule unique/méthodes , Sérine-thréonine kinases TOR/métabolisme ,
7.
Lancet Haematol ; 4(12): e595-e606, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-29153976

RÉSUMÉ

BACKGROUND: Patients with refractory or relapsed haematological malignancies have few treatment options and short survival times. Identification of effective therapies with genomic-based precision medicine is hampered by intratumour heterogeneity and incomplete understanding of the contribution of various mutations within specific cancer phenotypes. Ex-vivo drug-response profiling in patient biopsies might aid effective treatment identification; however, proof of its clinical utility is limited. METHODS: We investigated the feasibility and clinical impact of multiparametric, single-cell, drug-response profiling in patient biopsies by immunofluorescence, automated microscopy, and image analysis, an approach we call pharmacoscopy. First, the ability of pharmacoscopy to separate responders from non-responders was evaluated retrospectively for a cohort of 20 newly diagnosed and previously untreated patients with acute myeloid leukaemia. Next, 48 patients with aggressive haematological malignancies were prospectively evaluated for pharmacoscopy-guided treatment, of whom 17 could receive the treatment. The primary endpoint was progression-free survival in pharmacoscopy-treated patients, as compared with their own progression-free survival for the most recent regimen on which they had progressive disease. This trial is ongoing and registered with ClinicalTrials.gov, number NCT03096821. FINDINGS: Pharmacoscopy retrospectively predicted the clinical response of 20 acute myeloid leukaemia patients to initial therapy with 88·1% accuracy. In this interim analysis, 15 (88%) of 17 patients receiving pharmacoscopy-guided treatment had an overall response compared with four (24%) of 17 patients with their most recent regimen (odds ratio 24·38 [95% CI 3·99-125·4], p=0·0013). 12 (71%) of 17 patients had a progression-free survival ratio of 1·3 or higher, and median progression-free survival increased by four times, from 5·7 (95% CI 4·1-12·1) weeks to 22·6 (7·4-34·0) weeks (hazard ratio 3·14 [95% CI 1·37-7·22], p=0·0075). INTERPRETATION: Routine clinical integration of pharmacoscopy for treatment selection is technically feasible, and led to improved treatment of patients with aggressive refractory haematological malignancies in an initial patient cohort, warranting further investigation. FUNDING: Austrian Academy of Sciences; European Research Council; Austrian Science Fund; Austrian Federal Ministry of Science, Research and Economy; National Foundation for Research, Technology and Development; Anniversary Fund of the Austrian National Bank; MPN Research Foundation; European Molecular Biology Organization; and Swiss National Science Foundation.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs hématologiques/traitement médicamenteux , Adénine/analogues et dérivés , Adulte , Sujet âgé , Aire sous la courbe , Moelle osseuse/anatomopathologie , Bortézomib/usage thérapeutique , Chloro-2 désoxyadénosine/usage thérapeutique , Survie sans rechute , Femelle , Tumeurs hématologiques/imagerie diagnostique , Tumeurs hématologiques/mortalité , Tumeurs hématologiques/anatomopathologie , Humains , Estimation de Kaplan-Meier , Leucémie aigüe myéloïde/traitement médicamenteux , Mâle , Microscopie de fluorescence , Adulte d'âge moyen , Odds ratio , Projets pilotes , Pipéridines , Tomographie par émission de positons couplée à la tomodensitométrie , Pyrazoles/usage thérapeutique , Pyrimidines/usage thérapeutique , Courbe ROC , Induction de rémission , Jeune adulte
8.
Nat Chem Biol ; 13(6): 681-690, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-28437395

RÉSUMÉ

Small-molecule drugs may complement antibody-based therapies in an immune-oncology setting, yet systematic methods for the identification and characterization of the immunomodulatory properties of these entities are lacking. We surveyed the immumomodulatory potential of 1,402 small chemical molecules, as defined by their ability to alter the cell-cell interactions among peripheral mononuclear leukocytes ex vivo, using automated microscopy and population-wide single-cell image analysis. Unexpectedly, ∼10% of the agents tested affected these cell-cell interactions differentially. The results accurately recapitulated known immunomodulatory drug classes and revealed several clinically approved drugs that unexpectedly harbor the ability to modulate the immune system, which could potentially contribute to their physiological mechanism of action. For instance, the kinase inhibitor crizotinib promoted T cell interactions with monocytes, as well as with cancer cells, through inhibition of the receptor tyrosine kinase MSTR1 and subsequent upregulation of the expression of major histocompatibility complex molecules. The approach offers an attractive platform for the personalized identification and characterization of immunomodulatory therapeutics.


Sujet(s)
Immunomodulation/effets des médicaments et des substances chimiques , Bibliothèques de petites molécules/pharmacologie , Myosines cardiaques/métabolisme , Lignée cellulaire tumorale , Tumeurs du côlon/traitement médicamenteux , Crizotinib , Humains , Chaînes lourdes de myosine/métabolisme , Pyrazoles/pharmacologie , Pyridines/pharmacologie
9.
J Nucl Med ; 57(6): 943-9, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-26912427

RÉSUMÉ

UNLABELLED: Small organic ligands, selective for tumor-associated antigens, are increasingly being considered as alternatives to monoclonal antibodies for the targeted delivery of diagnostic and therapeutic payloads such as radionuclides and drugs into neoplastic masses. We have previously described a novel acetazolamide derivative, a carbonic anhydrase ligand with high affinity for the tumor-associated isoform IX (CAIX), which can transport highly potent cytotoxic drugs into CAIX-expressing solid tumors. The aim of the present study was to quantitatively investigate the biodistribution properties of said ligand and understand whether acetazolamide conjugates merit further development as drug carriers and radioimaging agents. METHODS: The conjugate described in this study, consisting of a derivative of acetazolamide, a spacer, and a peptidic (99m)Tc chelator, was labeled using sodium pertechnetate under reducing conditions and injected intravenously into CAIX-expressing SKRC-52 xenograft-bearing mice. Animals were sacrificed, and organ uptake as percentage injected activity of radiolabeled ligand per gram of tissues (%IA/g) was evaluated between 10 min and 24 h. Additionally, postmortem imaging by SPECT was performed. RESULTS: The acetazolamide conjugate described in this study could be labeled to high radiochemical purity (>95%, 2.2-4.5 MBq/nmol). Analysis of organ uptake at various time points revealed that the ligand displayed a maximal tumor accumulation 3 h after intravenous injection (22 %IA/g), with an excellent tumor-to-blood ratio of 70:1 at the same time point. The ligand accumulation in the tumor was more efficient than in any other organ, but a residual uptake in the kidney, lung, and stomach (9, 16, and 10 %IA/g, respectively) was also observed, in line with patterns of carbonic anhydrase isoform expression in those tissues. Interestingly, tumor-to-organ ratios improved on administration of higher doses of radiolabeled ligand, suggesting that certain binding sites in normal organs can be saturated in vivo. CONCLUSION: The (99m)Tc-labeled acetazolamide conjugate exhibits high tumor uptake and favorable tumor-to-kidney ratios of up to 3 that may allow imaging of tumors in the kidney and distant sites at earlier time points than commonly possible with antibody-based products. These data suggest that the described molecule merit further development as a radioimaging agent for CAIX-expressing renal cell carcinoma.


Sujet(s)
Carbonic anhydrase IX/métabolisme , Néphrocarcinome/métabolisme , Tumeurs du rein/métabolisme , Technétium/composition chimique , Acétazolamide/composition chimique , Acétazolamide/métabolisme , Acétazolamide/pharmacocinétique , Animaux , Néphrocarcinome/imagerie diagnostique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Humains , Marquage isotopique , Tumeurs du rein/imagerie diagnostique , Ligands , Souris , Distribution tissulaire , Tomographie par émission monophotonique
10.
Nat Chem ; 8(2): 103-13, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26791892

RÉSUMÉ

Nature has produced intricate machinery to covalently diversify the structure of proteins after their synthesis in the ribosome. In an attempt to mimic nature, chemists have developed a large set of reactions that enable post-expression modification of proteins at pre-determined sites. These reactions are now used to selectively install particular modifications on proteins for many biological and therapeutic applications. For example, they provide an opportunity to install post-translational modifications on proteins to determine their exact biological roles. Labelling of proteins in live cells with fluorescent dyes allows protein uptake and intracellular trafficking to be tracked and also enables physiological parameters to be measured optically. Through the conjugation of potent cytotoxicants to antibodies, novel anti-cancer drugs with improved efficacy and reduced side effects may be obtained. In this Perspective, we highlight the most exciting current and future applications of chemical site-selective protein modification and consider which hurdles still need to be overcome for more widespread use.


Sujet(s)
Découverte de médicament/méthodes , Protéines/composition chimique , Humains
11.
Nat Chem ; 7(3): 241-9, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25698334

RÉSUMÉ

In contrast to standard fragment-based drug discovery approaches, dual-display DNA-encoded chemical libraries have the potential to identify fragment pairs that bind simultaneously and benefit from the chelate effect. However, the technology has been limited by the difficulty in unambiguously decoding the ligand pairs from large combinatorial libraries. Here we report a strategy that overcomes this limitation and enables the efficient identification of ligand pairs that bind to a target protein. Small organic molecules were conjugated to the 5' and 3' ends of complementary DNA strands that contain a unique identifying code. DNA hybridization followed by an inter-strand code-transfer created a stable dual-display DNA-encoded chemical library of 111,100 members. Using this approach we report the discovery of a low micromolar binder to alpha-1-acid glycoprotein and the affinity maturation of a ligand to carbonic anhydrase IX, an established marker of renal cell carcinoma. The newly discovered subnanomolar carbonic anhydrase IX binder dramatically improved tumour targeting performance in vivo.


Sujet(s)
Découverte de médicament , Bibliothèques de petites molécules , Animaux , Carbonic anhydrases/composition chimique , ADN/composition chimique , Horseradish peroxidase/composition chimique , Ligands , Souris , Souris de lignée BALB C , Souris nude , Hybridation d'acides nucléiques , Orosomucoïde/composition chimique , Streptavidine/composition chimique
12.
Curr Opin Chem Biol ; 26: 48-54, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25721398

RÉSUMÉ

Carbonic anhydrase IX (CAIX) is expressed in many solid tumors in response to hypoxia and plays an important role in tumor acid-base homeostasis under these conditions. It is also constitutively expressed in the majority of renal cell carcinoma. Its functional inhibition with small molecules has recently been shown to retard tumor growth in murine models of cancer, reduce metastasis and tumor stem cell expansion. Additionally, CAIX is a promising antigen for targeted drug delivery approaches. Initially validated with anti-CAIX antibodies, the tumor-homing capacity of high-affinity small-molecule ligands of CAIX has recently been demonstrated. Indeed, conjugates formed of CAIX ligands and potent cytotoxic drugs could eradicate CAIX-expressing solid tumors in mice. These results suggest that CAIX is a promising target for the development of novel therapies for the treatment of solid tumors.


Sujet(s)
Acétazolamide/pharmacologie , Antigènes néoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Inhibiteurs de l'anhydrase carbonique/pharmacologie , Carbonic anhydrases/composition chimique , Tumeurs colorectales/traitement médicamenteux , Bibliothèques de petites molécules/pharmacologie , Acétazolamide/analogues et dérivés , Acétazolamide/synthèse chimique , Acétazolamide/métabolisme , Animaux , Antigènes néoplasiques/métabolisme , Antinéoplasiques/synthèse chimique , Antinéoplasiques/métabolisme , Carbonic anhydrase IX , Inhibiteurs de l'anhydrase carbonique/synthèse chimique , Inhibiteurs de l'anhydrase carbonique/métabolisme , Carbonic anhydrases/métabolisme , Lignée cellulaire tumorale , Tumeurs colorectales/enzymologie , Tumeurs colorectales/anatomopathologie , Humains , Ligands , Souris , Thérapie moléculaire ciblée , Multimérisation de protéines , Bibliothèques de petites molécules/synthèse chimique , Bibliothèques de petites molécules/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Angew Chem Int Ed Engl ; 53(16): 4231-5, 2014 Apr 14.
Article de Anglais | MEDLINE | ID: mdl-24623670

RÉSUMÉ

Antibody-drug conjugates are a very promising class of new anticancer agents, but the use of small-molecule ligands for the targeted delivery of cytotoxic drugs into solid tumors is less well established. Here, we describe the first small-molecule drug conjugates for the treatment of carbonic anhydrase IX expressing solid tumors. Using ligand-dye conjugates we demonstrate that such molecules can preferentially accumulate inside antigen-positive lesions, have fast targeting kinetics and good tumor-penetrating properties, and are easily accessible by total synthesis. A disulfide-linked drug conjugate with the maytansinoid DM1 as the cytotoxic payload and a derivative of acetazolamide as the targeting ligand exhibited a potent antitumor effect in SKRC52 renal cell carcinoma in vivo. It was furthermore superior to sunitinib and sorafenib, both small-molecule standard-of-care drugs for the treatment of kidney cancer.


Sujet(s)
Carbonic anhydrases/composition chimique , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/thérapie , Animaux , Systèmes de délivrance de médicaments , Humains , Souris , Structure moléculaire , Promédicaments
14.
Cancer Res ; 74(9): 2569-78, 2014 May 01.
Article de Anglais | MEDLINE | ID: mdl-24520075

RÉSUMÉ

It is generally thought that the anticancer efficacy of antibody-drug conjugates (ADC) relies on their internalization by cancer cells. However, recent work on an ADC that targets fibronectin in the tumor microenvironment suggests this may not be necessary. The alternatively spliced extra domains A and B (EDA and EDB) of fibronectin offer appealing targets for ADC development, because the antigen is strongly expressed in many solid human tumors and nearly undetectable in normal tissues except for the female reproductive system. In this study, we describe the properties of a set of ADCs based on an antibody targeting the alternatively spliced EDA of fibronectin coupled to one of a set of potent cytotoxic drugs (DM1 or one of two duocarmycin derivatives). The DM1 conjugate SIP(F8)-SS-DM1 mediated potent antitumor activity in mice bearing DM1-sensitive F9 tumors but not DM1-insensitive CT26 tumors. Quantitative biodistribution studies and microscopic analyses confirmed a preferential accumulation of SIP(F8)-SS-DM1 in the subendothelial extracellular matrix of tumors, similar to the pattern observed for unmodified antibody. Notably, we found that treatments were well tolerated at efficacious doses that were fully curative and compatible with pharmaceutical development. Our findings offer a preclinical proof-of-concept for curative ADC targeting the tumor microenvironment that do not rely upon antigen internalization.


Sujet(s)
Anticorps/pharmacologie , Antinéoplasiques/pharmacologie , Maitansine/pharmacologie , Animaux , Anticorps/usage thérapeutique , Antinéoplasiques/pharmacocinétique , Antinéoplasiques/usage thérapeutique , Cellules CHO , Lignée cellulaire tumorale , Cricetinae , Cricetulus , Cellules souches de carcinome embryonnaire , Femelle , Humains , Maitansine/pharmacocinétique , Maitansine/usage thérapeutique , Souris , Souris de souche-129 , Souris de lignée BALB C , Souris nude , Distribution tissulaire , Tests d'activité antitumorale sur modèle de xénogreffe
15.
J Proteomics ; 99: 138-51, 2014 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-24487095

RÉSUMÉ

A detailed characterization of the cell surface proteome facilitates the identification of target antigens, which can be used for the development of antibody-based therapeutics for the treatment of hematological malignancies. We have performed cell surface biotinylation of five human myeloid leukemia cell lines and normal human granulocytes, which was used for mass spectrometric analysis and allowed the identification and label-free, relative quantification of 320 membrane proteins. Several proteins exhibited a pronounced difference in expression between leukemia cell lines and granulocytes. We focused our attention on CD166/ALCAM, as this protein was strongly up-regulated on all AML cell lines and AML blasts of some patients. A human monoclonal antibody specific to CD166 (named H8) was generated using phage display technology. H8 specifically recognized AML cells in FACS analysis while demonstrating tumor targeting properties in vivo. After in vitro screening of five potent cytotoxic agents, a duocarmycin derivative was used for the preparation of an antibody-drug conjugate, which was able to kill AML cells in vitro with an IC50 of 8nM. The presented atlas of surface proteins in myeloid leukemia provides an experimental basis for the choice of target antigens, which may be used for the development of anti-AML therapeutic antibodies. BIOLOGICAL SIGNIFICANCE: The ability to discriminate between malignant and healthy, essential cells represents an important requirement for the development of armed antibodies for the therapy of hematological malignancies. Our proteomic study is, to our knowledge, the first large scale comparison of the accessible cell surface proteome of leukemia cells and normal blood cells, facilitating the choice of a suitable target for the treatment of acute myeloid leukemia (AML). An antibody drug conjugate was generated recognizing the CD166 antigen which was found to be strongly up-regulated in all AML cell lines and AML blasts of some patients. This antibody drug conjugate SIP(H8)-Duo might be further characterized in therapy experiments and might lead to a new targeted treatment option for AML.


Sujet(s)
Anticorps monoclonaux/immunologie , Anticorps antitumoraux/immunologie , Antigènes CD/immunologie , Molécules d'adhérence cellulaire neuronale/immunologie , Protéines foetales/immunologie , Leucémie aigüe myéloïde/immunologie , Protéines tumorales/immunologie , Protéome/immunologie , Animaux , Anticorps monoclonaux/usage thérapeutique , Anticorps antitumoraux/usage thérapeutique , Antigènes CD/métabolisme , Molécules d'adhérence cellulaire neuronale/métabolisme , Protéines foetales/métabolisme , Cellules HL-60 , Humains , Cellules K562 , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris de lignée BALB C , Souris nude , Protéines tumorales/métabolisme , Protéome/métabolisme , Protéomique/méthodes
16.
Bioconjug Chem ; 24(2): 234-41, 2013 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-23350721

RÉSUMÉ

Pretargeting approaches rely on the injection of bispecific antibodies capable of recognizing both an accessible disease marker and a small ligand, which is typically administered at a later stage and which serves as delivery vehicle for a payload for imaging or therapy applications. In the oncology field, pretargeting strategies have exhibited extremely promising biodistribution results and in vivo selectivity, but have often relied on the cumbersome preparation of multispecific antibodies by chemical conjugation techniques. Here, we describe the design, production, and characterization of a novel class of bispecific multivalent antibody products, which contain both antibody fragments and an anticalin moiety for the simultaneous recognition of tumor-associated antigens and a small organic molecule. Anticalins are derivatives of the naturally occurring binding proteins lipocalins, which have been engineered to recognize a target molecule with high affinity. In particular, we produced and compared in vitro and in vivo different fusion proteins, which contained the anticalin FluA that selectively recognizes various different fluorescein derivatives and the F8 antibody specific to the alternatively spliced EDA domain of fibronectin (a marker of tumor angiogenesis). The selective accumulation of the most promising fusion-protein scFv(F8)-FluA-scFv(F8) on solid tumors and simultaneous binding of fluorescein derivatives could be visualized in vivo using a fluorescein-near-infrared fluorescent dye conjugate, confirming the potential of antibody-anticalin fusion proteins for pretargeting applications.


Sujet(s)
Anticorps bispécifiques , Antigènes néoplasiques/analyse , Fibronectines/analyse , Lipocalines , Tumeurs/diagnostic , Protéines de fusion recombinantes , Protéines recombinantes , Anticorps à chaîne unique , Animaux , Anticorps bispécifiques/génétique , Anticorps bispécifiques/immunologie , Antigènes néoplasiques/immunologie , Cellules CHO , Clonage moléculaire , Cricetinae , Fibronectines/immunologie , Lipocalines/génétique , Lipocalines/immunologie , Souris , Souris de lignée BALB C , Souris nude , Tumeurs/immunologie , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie
17.
Angew Chem Int Ed Engl ; 52(5): 1384-402, 2013 Jan 28.
Article de Anglais | MEDLINE | ID: mdl-23296451

RÉSUMÉ

The targeted delivery of potent cytotoxic agents has emerged as a promising strategy for the treatment of cancer and other serious conditions. Traditionally, antibodies against markers of disease have been used as drug-delivery vehicles. More recently, lower molecular weight ligands have been proposed for the generation of a novel class of targeted cytotoxics with improved properties. Advances in this field crucially rely on efficient methods for the identification and optimization of organic molecules capable of high-affinity binding and selective recognition of target proteins. The advent of DNA-encoded chemical libraries allows the construction and screening of compound collections of unprecedented size. In this Review, we survey developments in the field of small ligand-based targeted cytotoxics and show how innovative library technologies will help develop the drugs of the future.


Sujet(s)
ADN/composition chimique , Vecteurs de médicaments/composition chimique , Bibliothèques de petites molécules/composition chimique , Anticorps/composition chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/toxicité , ADN/métabolisme , Humains , Ligands , Peptides/composition chimique , Vitamines/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...