ANTI-HER2 POLYPEPTIDES DERIVATIVES AS NEW DIAGNOSTIC MOLECULAR PROBES

Information

  • Patent Application
  • 20230040008
  • Publication Number
    20230040008
  • Date Filed
    December 16, 2020
    3 years ago
  • Date Published
    February 09, 2023
    a year ago
Abstract
The present invention provides new polypeptide derivatives binding to Human Epidermal Growth Factor Receptor 2 (HER2) and their conjugates thereof, and to their use as a diagnostic agent, particularly for early detection, patient stratification and treatment monitoring of forms of cancer characterized by over-expression of HER2.
Description

The present invention relates to new polypeptide derivatives which bind to Human Epidermal Growth Factor Receptor 2 (HER2) and their conjugates thereof, and to their use as diagnostic agents, particularly for early detection, patient stratification and treatment monitoring of forms of cancer characterized by over-expression of HER2.


BACKGROUND

In recent years biomarkers discovery is emerging as a dynamic and powerful approach for its potential in identifying the earliest events for a pathological state, both in diagnostic and therapeutic fields. It is also becoming extremely challenging to develop probes able to recognize a target involved in a molecular pathway of a disease state. These probes have to be able to improve the diagnostic procedures for better application of current therapeutic regimens, improving the therapy efficacy and reducing patients overtreatments.


The proto-oncogene HER2 or HER2/neu (Human Epidermal Growth Factor Receptor 2) encodes the production of a cell surface receptor, known as HER2 protein or receptor, a member of ErbB family of tyrosine kinase receptors involved in transmission of signals controlling normal cell growth and differentiation. Due to errors in the DNA replication system in tumor cells, HER2 gene can be amplified leading to an over-expression of the HER2 protein on the cells surface. In particular, over-expression of the HER2 receptor occurs in 25-30% of human breast primary carcinomas1 and has been described in correlation with many other HER2 positive tumors, including for instance head&neck cancer, ovarian cancer, lung cancer, bladder cancer and tumors of the gastrointestinal tract.


For these reasons, HER2 represents an important molecular target for many oncological therapies and diagnostic methods. Among the approaches to inhibit HER2 protein on tumor cell surfaces, some therapies using humanized variants of monoclonal antibodies, e.g. trastuzumab and pertuzumab, have become commercially available in recent years for the treatment of HER2-positive tumors; however, despite the clinical success, their use is associated with some toxic effects and undesired side-effects on healthy tissues which are a reason of concern.


In general, many antibodies are characterized by slow and inefficient tumor penetration, long biodistribution time and slow clearance from the blood, and, for diagnostic applications, require the use of long-lived radioisotopes and imaging at late time points, thus exposing patients to a high radiation burden. Additionally, monoclonal antibodies may be immunogenic, thus precluding repeated administration for routine diagnostic procedures.


Therefore, there is still a high need to a continued provision of new molecules able to interact with HER2 forms.


These problems could be circumvented for instance by the use of small molecules.


Recently, small binding polypeptides (4 to 20 kDa), such as for instance affibodies, fibronectins and DARPins, have been described and are attracting interest as potential alternatives to antibodies for use in non-invasive imaging methods. Generally, such peptides have demonstrated to be biocompatible and poorly immunogenic in vivo, highly selective and able to bind and recognize specific targets, and present an affinity constant (Kd) in the nanomolar range, lower than the antibodies, the latters generally having a Kd in the milli/micromolar range. Also, small polypeptides penetrate tissues faster and more efficiently because of their significantly lower molecular weight and can distinguish extracellular or intracellular domains of proteins, which cannot be differentiated by antibodies.


Moreover, unlike antibodies and other large macromolecules (Mw˜150 kDa), small molecules clear rapidly from circulation and thus reach at early time-points after administration a tumor/blood ratio suitable for imaging. This in turn allows physicians to obtain diagnostic information much more quickly than with antibody-based imaging agents.


In particular, the development of new small imaging probes binding HER2 receptor can be of great value providing specific imaging agents for patient stratification, for predicting or monitoring the response to specific anti-cancer therapies and even in the image-guided surgery of the tumors by means of optical imaging, magnetic resonance imaging, nuclear imaging, computed tomography imaging, ultrasound and multimodality imaging techniques.


In particular, among the class of antibody-mimetic molecules, affitins are compounds consisting of small, single-chain affinity proteins (7 kDa, generally 66 amino acids) which are being studied for their high tissue penetration potential. Known examples of such peptides are Nanofitins® (Affilogic S.A.S., France). These polypeptides are derived from the hyperstable DNA-binding protein Sac7d of the thermophilic archaeon Sulfolobus acidocaldarius, from which they retain most of the advantageous biophysical features, such as resistance to temperature (up to 90° C.) and pH (0-13). In particular, high-affinity affitins are engineered with several rounds of ribosome display by the full randomization of 10-14 amino acid residues in the DNA-binding site of Sac7d and can be tuned to have high affinity towards specific targets expressed on the surface of cancer cells, while conserving the original stability features of being thermophilic and acidophilic2,3.


Moreover, besides being extremely stable and robust, affitin polypeptides have better pharmacokinetic profiles for imaging with respect to antibodies and can easily produced in large quantities employing recombinant bacterial technologies.


For instance, Goux M. et al., Bioconjugated Chem. 2017, 28, 2361-23714 discloses the use of the anti-EGFR Nanofitin B10 as imaging agent, in particular as a target PET radiotracer, when radiolabeled with 18F-FBEM. However, no specific sequences of affitins binding the target HER2 have been described yet.


Among the known antibody-mimetic polypeptides, several sequences of affibodies binding to HER2 have been investigated, such as those described in WO2009/0808105 in the name of Affibody AB and in Orlova A. et al., Cancer Res. 2006, 66(8), 4339-43486.


An example of application of such HER2-binding polypeptides as imaging agents after conjugation with a radionuclide and a chelator is reported in WO2012/0967607 in the name of GE Healthcare Ltd and Affibody AB.


WO2017/1610968, in the name of Tarveda Therapeutics Inc., discloses nano- and micro-particles comprising generic conjugates with a targeting moiety, such as for instance a Nanofitin, a linker and an active agent, even though without reporting any specific example of such conjugates.


Therefore, despite the efforts, there is still the need to identify and develop HER2 binding agents as described above, characterized by a high and specific affinity for the target, to be used as diagnostic agents of HER2-positive tumors, in particular for monitoring the treatments of HER2-positive tumors.


SUMMARY

The present invention relates to new polypeptides sequences, characterized by the affitins structure, specifically targeting HER2 receptor with high affinity.


In particular the invention relates to HER2-binding polypeptides comprising the amino acid sequence:









(SEQ ID NO: 1)


VKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGKAGPGRVPEKDAP





KELLDMLARAEREK.






Considering the overexpression of HER2 protein as a marker of pathological states, being involved in breast cancer and in many other types of tumor, the HER2-binding affitins of the invention may be useful, once labelled with an imaging moiety, for an early cancer diagnosis and staging in the perspective of more effective therapeutic treatments and/or of reducing patients overtreatments.


To this purpose, they can be efficiently conjugated to labelling moieties, allowing an easy recognition and evaluation during in vitro and/or in vivo analyses. Such conjugates or complexes are also object of the present invention.


The affitins of the invention were found endowed with properties suitable for their use in diagnostic tools, such as a nanomolar affinity for the target HER2, absence of binding to albumins, low predicted immunogenicity, good internalization in HER2-positive cells and a good expression yield. In particular, the affitins of the invention and their conjugates are able to specifically recognize the target at the site where it is physiologically present, i.e. on the cell surface where HER2 is expressed, thus allowing possible in vivo applications thereof.


Moreover, it has been found that the affitins of the invention are able to recognize the target HER2 at a specific epitope distinct from that recognized by the monoclonal antibodies trastuzumab and pertuzumab, currently used in clinical treatment of HER2-positive tumors, thus allowing for instance the use of the affitins of the invention as agents to test for changes in HER2 receptor expression in response to trastuzumab and/or pertuzumab therapy.





DESCRIPTION OF THE DRAWINGS

The features of the invention can be better understood with reference to the following detailed description and the accompanying figures, wherein:



FIG. 1 represents a SE-HPLC chromatogram of the purified IRDye800CW-Aff02 conjugate;



FIG. 2 reports the results of a competitive ELISA test with trastuzumab-biotin or pertuzumab-biotin vs Aff02 polypeptide;



FIG. 3 reports the results of a different competitive ELISA test with trastuzumab-biotin vs Aff02 polypeptide (a) or pertuzumab-biotin vs Aff02 polypeptide (b);



FIG. 4 reports the curves of trastuzumab (a) or pertuzumab (b) in the presence or absence of Aff02;



FIG. 5 reports the fluorescence signal curves of IRDye800CW-Aff02 conjugate administered at a dosage of 10 nmol/mouse in Balb/c nu/nu mice (Mean, St. Dev, n=3);



FIG. 6 reports the ex-vivo biodistribution of conjugate IRDye800CW-Aff02 in healthy mice at 48 h after injection at a dosage 10 nmol/mouse.





BRIEF DESCRIPTION OF THE SEQUENCE LISTING





    • SEQ ID NO: 1 sets out the 64 aac sequence of the affitin of the invention named Aff01:












VKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGKAGPGRVPEKDAP





KELLDMLARAEREK;








    • SEQ ID NO: 2 sets out the 77 aac sequence corresponding to a derivatized variant of SEQ ID NO: 1, engineered with a Cys at the C-terminus and a hexahistidine tag at the N-terminus, named affitin Aff02:












MRGSHHHHHHGSVKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGK





AGPGRVPEKDAPKELLDMLARAEREKC;








    • SEQ ID NO: 3 identifies the His6-tag derivative linked to the N-terminus of the affitin Aff02 and Aff02-0:














MRGSHHHHHHGS;








    • SEQ ID NO: 4 is an engineered polypeptide corresponding to Aff02 (SEQ ID NO: 2) without the C-terminal cysteine and is named Aff02-0:












MRGSHHHHHHGSVKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGK





AGPGRVPEKDAPKELLDMLARAEREK;








    • SEQ ID NO: 5 identifies the engineered polypeptide formed by fusion of the sequence Aff02-0 (SEQ ID NO: 4) with a truncated portion of Pseudomonas exotoxin A (Pe38):












MRGSHHHHHHGSVKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGK





AGPGRVPEKDAPKELLDMLARAEREKKLGSAGSAAGSGEFGGSLAALTAH





QACHLPLETFTRHRQPRGWEQLEQCGYPVQRLVALYLAARLSWNQVDQVI





RNALASPGSGGDLGEAIREQPEQARLALTLAAAESERFVRQGTGNDEAGA





ASGPADSGDALLERNYPTGAEFLGDGGDVSFSTRGTQNWTVERLLQAHRQ





LEERGYVFVGYHGTFLEAAQSIVFGGVRARSQDLDAIWRGFYIAGDPALA





YGYAQDQEPDARGRIRNGALLRVYVPRSSLPGFYRTGLTLAAPEAAGEVE





RLIGHPLPLRLDAITGPEEEGGRLETILGWPLAERTVVIPSAIPTDPRNV





GGDLDPSSIPDKEQAISALPDYASQPGKPPKDEL.






DETAILED DESCRIPTION

It is an object of the present invention to provide new agents with affinity for Human Epidermal Growth Factor Receptor 2 (HER2), in particular a polypeptide that is characterized by specific binding to HER2 and its derivatives and conjugates thereof.


Thus, in a first aspect, the invention provides a polypeptide that specifically binds to HER2 comprising the amino acid sequence VKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGKAGPGRVPEKDAPKELLD MLARAEREK (SEQ ID NO: 1). Preferably, such sequence has a length of up to 100 amino acids. More preferably, it has a length of up to 80 amino acids.


In a preferred specific embodiment, the invention provides the polypeptide consisting of the amino acid sequence as set out in SEQ ID NO: 1.


Different modifications of, and/or addition to, the polypeptides above defined may be performed, without departing from the scope of the present invention, including for instance addition of further amino acids to the sequence, derivatization with any linker or spacer, or conjugation with specific labels or imaging moieties, as described in more detail below.


For instance, the invention also encompasses polypeptides in which the HER2 binding polypeptide described above bears additional amino acid residues added at one or both terminal positions, provided that they not alter the biological function of said peptides. These additional amino acid residues may play a role in the binding of HER2 by the polypeptide, but may equally well serve other purposes, related for example to the production, purification, stabilization, coupling and/or detection of the polypeptide. Preferably, such additional amino acid residues may comprise one or more amino acid residues added for purposes of chemical coupling. A preferred embodiment relates to the addition at the very first or very last position in the polypeptide chain, i.e. at the N or C terminus, of at least one amino acid containing a thiol group, such as cysteine or homocysteine, or containing in the side chain a primary amino group, such as lysine, or a carboxylic acid group, such as glutamic acid or aspartic acid.


In another embodiment, such residue to be used for chemical coupling may also be introduced by replacement of another amino acid on the surface of the protein domain, preferably on a portion of the surface that is not involved in target binding.


In some embodiments, said residue suitable for the coupling can also be represented by a synthetic or unnatural amino acid or an amino acid mimetic that function similarly to the natural occurring amino acids. Non-limiting examples are selected from β2- or β3-amino acids; γ-amino acids; substituted glycine or alanine, such as p-acetophenylalanine or p-azidophenylalanine; 6-amino hexanoic acid and other derivatives known in the art, which are useful to modulate the coupling strategy or to confer stability to the conjugate.


The additional amino acid residues may also comprise a “tag” useful for the purification, isolation or detection of the polypeptide, such as for instance a hexahistidyl tag, or a “myc” tag or a “FLAG” tag for interaction with antibodies specific to the tag, or other alternatives well known to the skilled person, which can be alone or coupled to a binding target.


Accordingly, in another preferred embodiment the invention provides a HER2 binding polypeptide comprising the amino acid sequence MRGSHHHHHHGSVKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGKAGPGR VPEKDAPKELLDMLARAEREKC (SEQ ID NO: 2), corresponding to the peptide of SEQ ID NO: 1 modified by addition of a Cys at the C-terminus and of the His6-tag of amino acid sequence MRGSHHHHHHGS (SEQ ID NO: 3) at the N-terminus.


In a preferred embodiment the invention provides the HER2 binding polypeptide consisting of the amino acid sequence as set out in SEQ ID NO: 2.


In another preferred embodiment the invention provides a HER2 binding polypeptide comprising the amino acid sequence MRGSHHHHHHGSVKVKFWGAGVEKEVDTSKITWVTRSGKYVIFTYDDNGKAGPGR VPEKDAPKELLDMLARAEREK (SEQ ID NO: 4), corresponding to the peptide of SEQ ID NO: 1 modified by addition of the His6-tag of amino acid sequence MRGSHHHHHHGS (SEQ ID NO: 3) at the N-terminus.


In another embodiment the invention provides the HER2 binding polypeptide consisting of the amino acid sequence as set out in SEQ ID NO: 4.


In particular, the additional hexahistidine-tag at the N-terminus of the peptide is useful for purification of the protein by Ni-NTA column, following known procedures, as described for instance in Bornhorst J. A. et al, Methods Enzymol. 2000, 326, 245-2549.


The invention also encompasses multimers of the polypeptide, such as a dimer, comprising sequence SEQ ID NO: 1. For instance, in one embodiment the invention provides for a homodimer molecule including two polypeptides comprising amino acid sequence SEQ ID NO: 1 or a heterodimer molecule including a polypeptide comprising amino acid sequence SEQ ID NO: 1 and a different polypeptide with high binding affinity for HER2 or for other target molecules, in order to create multispecific reagents that may be used in several biotechnological applications.


The linked polypeptide “units” in such multimers according to the invention may be connected by covalent coupling using known organic chemistry methods, or expressed as one or more fusion polypeptides in a system for recombinant expression of polypeptides, or joined in any other fashion, either directly or via a linker, for example an amino acid linker.


All the above defined polypeptides can be considered as a suitable alternative to antibodies against HER2 in several applications. An aspect of the invention thus relates to a HER2 binding polypeptide as described above which is conjugated to a compound or labeled to form a reporter moiety amenable to be used in diagnosis or imaging of a cancer disease caused by and/or associated with an overexpression of HER2.


A “reporter moiety” refers to a molecule that can be directly or indirectly detected by an imaging technique, wherein the HER2 binding polypeptides of the invention are linked to at least one detectable label or material (e.g. a dye whose optical properties can be measured; a contrast agent comprising a magnetic particle; or a gas containing vesicle). A reporter moiety is undirectly detectable for instance when it becomes detectable only through interaction with an environment or a further material modifying its detectability.


Suitable imaging techniques for detection of such reporter moieties include for instance magnetic resonance, positron-emission tomography (PET), computed tomography (CT), ultrasounds (US), photoacoustic imaging (PAI), near-infrared fluorescence (NTRF) and single photon emission computed tomography (SPECT) or techniques correlated with optical imaging (OI).


In particular, PET imaging techniques also include immunoPET, wherein a specific biomarker information on a disease is obtained by directly targeting the receptor of interest, for instance with antibodies or antibody-mimetic molecules.


For imaging applications, the polypeptides of the invention are linked to a label generally selected from: a fluorophore moiety capable of generating a fluorescent signal, such as fluorescein, FITC, Alexa dyes, Cyanine dyes, DyLight dyes, IRDye dyes or VivoTag dyes; an optical moiety, including agents that may be used to produce contrast or signal using optical imaging; a magnetic or paramagnetic moiety, including a chelating agent for magnetic resonance which is able to form stable complexes with paramagnetic metal ions, including for instance Gd(III), Mn(II), Cr(III), Cu(II), Fe(III), Pr(III), Nd(III), Sm(III), Tb(III), Yb(III), Dy(III), Ho(III) and Er(III); a radiolabeled isotope, including for instance 18F, 124I, 11C, 64Cu, 68Ga, 89Zr, 44Sc and 99mTc and other radioisotopes of indium, gallium, yttrium, bismuth, radioactinides and radiolanthanides; an affinity label, such as biotin; an X-ray responsive moiety that may be used to produce contrast or signal using X-ray imaging, such as iodinated organic molecules or chelates of heavy metal ions; an ultrasound responsive moiety or assembly for contrast enhanced ultrasound imaging, preferably in the form of a gas-filled microbubble; a photoacoustic responsive imaging moiety, including photoacoustic imaging-compatible agents; and nanoparticle-based moieties.


Preferably, in case of labeling with radionuclides, such complexation is performed with the use of chelators or multidentate ligands, forming chelates in particular with the radiometals. Non-limiting examples of such chelators are 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA); 2,2′,2″-(1,4,7-triazacyclononane-1,4,7-triyl)triacetic acid (NOTA); 2-[bis[2-[bis(carboxymethyl)amino]ethyl]amino]acetic acid (DTPA); ethylenediaminetetraacetic acid (EDTA), 10-(2-hydroxypropyl)-1,4,7-tetraazacyclododecane-1,4,7-triacetic acid (HP-DO3A); 1,4-bis(carboxymethyl)-6-[bis(carboxymethyl)]amino-6-methylperhydro-1,4-diazepine (AAZTA) and derivatives thereof.


Accordingly, in a preferred embodiment the invention encompasses a radiolabeled polypeptide consisting of a chelate of a HER2 binding polypeptide described above and a radionuclide, such as a radioactive metal. More preferably, the radioactive metal is 68Ga, 44Sc or 99mTc.


In another preferred embodiment, the polypeptides of the invention are conjugated with a fluorophore moiety selected from the cyanine dyes, such as for instance IRDye800CW, IRDye650, IRDye680, Cy3, Cy5, Cy5.5, Cy7, ZW800-1, AlexaFluor 750 AlexaFluor 790, and analogues and derivatives thereof.


Moreover, the polypeptide and the label may be linked using a tag system, including biotin/avidin, biotin/streptavidin and biotin/NeutrAvidin.


Accordingly, in a further preferred aspect, the invention provides for polypeptides as defined above which are linked to an affinity label, such as biotin.


The coupling between the targeting polypeptide of the invention and the label or imaging moiety can be performed through methods described herein in the examples or other methods well known to the skilled person. For instance they may be linked either covalently or noncovalently, optionally by interposition of a suitable linker or spacer, useful for labeling.


Generally, such linkers or spacers may comprise amino acids or nucleic acids sequences or reactive moieties including for instance an aminoxy group, an azido group, an alkyne group, a thiol group or a maleimido group, either alone or in combination.


Such linkers preferably comprise two functional moieties, one providing rapid and efficient labeling and another enabling rapid and efficient coupling to the polypeptides of the invention, for instance through an amine group or preferably through the thiol group of the cysteine. For example a maleimide group reacts with thiol groups to form a stable thioether bond. Preferably, the final complex is formed by first reacting the label with the linker, and subsequently with the thiol group of the polypeptide.


The polypeptides of the invention, suitably conjugated with a label to form a reporter moiety, can be used in the diagnosis and staging of HER2-related states, disorders, dysfunctions, conditions or diseases and for monitoring of the treatment response in such states, particularly for targeting in the clinical setting of cancer diseases characterized by an overexpression of HER2 protein. Exemplary applications include the diagnosis of a cancer disease correlated with the expression of HER2, such as for instance breast cancer, head&neck cancer, ovarian cancer, lung cancer, bladder cancer and tumors of the gastrointestinal tract.


Accordingly, in a further aspect the invention provides a polypeptide conjugate as defined above for use in diagnosis or visualization of a cancer disease caused by and/or associated with an overexpression of HER2, i.e. in a process of identifying or determining the nature and cause of a HER2-correlated disease through evaluation of patient history, examination and review of laboratory data. In particular, such polypeptide conjugate may be used for imaging of a body tissue or organ system overexpressing HER2. More preferably, it can be used to determine HER2 expression before and after a treatment, which can be accomplished by obtaining images before and after the treatment, and to determine the extent or anatomical content of HER2 expression, e.g. for surgical purposes or for patients stratification.


In yet another aspect, the invention provides the use of a polypeptide conjugate as described above in the preparation of an imaging agent for detecting a cancer tissue or organ characterized by overexpression of HER2.


In a further aspect, the invention provides a composition comprising a polypeptide or polypeptide conjugate as herein defined and one or more suitable pharmaceutically acceptable carriers, excipients, diluents and/or additives. The ingredients of the composition can be varied depending on the intended use, whether for diagnostic or imaging applications. Such composition can also comprise one or more adjuvants selected from preservatives, wetting agents, emulsifying agents and dispersing agents.


The composition can further contain one or more different imaging agents.


In one embodiment, the invention provides the composition said above for use in the imaging of a target tissue bearing HER2, wherein the composition comprises a polypeptide of the invention conjugated or labeled with an imaging moiety as above defined. The composition can be e.g. also in the form of a liposome or nanoparticles and can be suitable for different types of administration. Preferably, said imaging is performed with an imaging technique selected from magnetic resonance, positron-emission tomography (PET), computed tomography (CT), ultrasounds (US), photoacoustic imaging (PAI), near-infrared fluorescence (NIRF) and single photon emission computed tomography (SPECT), or correlated with optical imaging (OI).


In one embodiment the composition is suitable for parenteral administration, preferably for intravenous or subcutaneous administration, for instance in the form of sterile aqueous solutions or dispersions or powders for the preparation of sterile injectable solutions, dispersions or emulsion formulations. In another embodiment the above composition may be administered topically or by inhalation.


Said compositions are provided for use with an imaging technique for visualizing HER2 expressing tumors, such as breast cancer.


In another aspect, the invention provides for a kit that contains at least one polypeptide of the invention, preferably labeled or conjugated with an imaging moiety, in one or more containers and the instructions for use.


The invention also provides for a method for an in vivo imaging of at least a portion of a mammalian subject, preferably a human, having a cancer characterized by overexpression of HER2, such method comprising the steps:

    • administering a composition as described above to the subject;
    • optionally monitoring the delivery of the composition to the subject;
    • imaging the subject with a diagnostic device; and
    • optionally diagnosing the subject with a HER-2-associated disease condition.


The polypeptides of the invention can be obtained by recombinant expression, i.e. by sequence cloning in an expression plasmid, which can be expressed for instance in Escherichia coli, and purification by affinity chromatography, for instance following the procedure described in Huet S. et al, PLoS ONE 2015, 10(11): e014230410.


For instance, precultures can be grown overnight at 37° C. in medium with glucose and antibiotics. Precultures can be diluted in medium with glucose and antibiotics, and grown at 37° C. to mid-log phase. Then, protein expression can be induced by addition of Isopropyl β-D-1-thiogalactopyranoside and the culture shaken at 30° C. overnight. Bacteria can be pelleted by centrifugation, then resuspended in a lysis buffer. Cell lysis can be performed at room temperature for 1 h and the suspension centrifuged to remove cell debris. Histag-proteins can then be purified from supernatants by immobilized metal ion affinity chromatography (IMAC), using Nickel resin and an elution buffer containing 250 mM Imidazole.


Another aspect of the present invention relates to a nucleic acid molecule which encodes a polypeptide as described above.


In a further embodiment the invention relates to an expression vector comprising the nucleic acid molecule said above and optionally other nucleic acid elements that enable production of the polypeptide according to the invention through expression of the nucleic acid molecule.


Moreover, the invention relates to a host cell (e.g. a eukaryotic, prokaryotic or plant cell) comprising said expression vector.


The above aspects represent recombinant techniques for the production of a polypeptide of the invention which are well known to the skilled person.


Alternatively, the polypeptides of the invention may also be produced by other known means, including chemical synthesis, for instance with standard solid phase synthesis techniques, or expression in different hosts, such as for instance plants and transgenic animals.


The invention will now be illustrated in detail through the description of examples performed in accordance therewith.


Experimental Section


The following examples are provided for illustration only and should not be construed as limiting the invention.


Equipment


The sequences and conjugates thereof of the present invention, as prepared according to the following examples, were characterized by UV/VIS (optical density), SE-HPLC and RP-HPLC analytical data, by use of one of the following methods:


SE-HPLC: The Size Exclusion HPLC analysis was carried out at 30° C. at a flow rate of 1 ml/min using a Sepax Zenix SEC-80 4.6×300 mm column (injection volume: 10 μL). The instrument was equipped with a UV/VIS detector at 280/780 nm. The mobile phase was 150 mM phosphate buffer, pH 7.0.


RP-HPLC: HPLC analyses were performed at 40° C. at a flow rate of 1.2 ml/min using a Jupiter Proteo (Phenomenex) 4.6×250 mm column (injection volume: 10 μL). The instrument was equipped with a UV/VIS detector at 280/780 nm. Mobile phase A was 0.1% TFA in water and mobile phase B was 0.1% TFA in acetonitrile. The gradient is reported below:












Gradient:









Min
% A
% B












0
72
28


2
72
28


8
65
35


9
10
90


11
10
90


12
72
28


17
72
28









Affinity experiments were performed by Biolayer Interferometry (BLI) assay, using an Octet System and Protein A biosensors (ForteBio). Recombinant human protein hHER2 was purchased from R&D Systems (Minneapolis, US).


In vivo imaging experiments were performed using the IVIS Spectrum In Vivo Imaging System (Perkin Elmer Inc.), equipped with with 10 narrow band excitation filters (30 nm bandwidth) and 18 narrow band emission filters (20 nm bandwidth) spanning 430-850 nm.


LIST OF ABBREVIATIONS

HER2 Human Epidermal Growth Factor Receptor 2


kDa kiloDalton


HSA Human serum albumin


MSA Mouse serum albumin


BSA Bovine serum albumin


ELISA Enzyme-linked immunosorbent assay


BLI Biolayer interferometry


Kd Dissociation equilibrium constant


aac Amino acid


RT Room temperature


Pe38 Truncated portion of Pseudomonas exotoxin A


TCEP Tris(2-carboxyethyl)phosphine


UV/VIS Ultraviolet/visible spectrophotometry


SE-HPLC Size exclusion high performance liquid chromatography


RP-HPLC Reversed-phase High Performance Liquid Chromatography


DTT Dithiothreitol


PBS Phosphate-buffered saline


TBS-T Tris-buffered saline—Tween 20


TMB Tetramethylbenzidine


TCEP Tris(2-carboxyethyl)phosphine


HRP Horseradish peroxidase


The abbreviations for individual amino acids residues are conventional: for example, Cys or C is cysteine, Asp or D is aspartic acid, Gly or G is glycine, Arg or R is arginine. The amino acids, as herein referred to, should be understood to be of the L-isomer configuration unless otherwise noted.


Example 1: Preparation of the Affitins According to the Invention

To selectively target HER2 receptor, a library of affitins has been enriched against the target by several consecutive rounds of screening (up to 6 rounds). The enrichment of anti-HER2 affitins has been followed after any selection round by ELISA assay monitoring, also controlling the condition of negligible binding to human (HSA), murine (MSA) and bovine (BSA) serum albumin. At the end of the selection process, the affitin named Aff01 (SEQ ID NO: 1) has been found endowed with nanomolar affinity to HER2, absence of binding to albumin and low in silico immunogenity.


The polypeptide Aff02 was obtained by sequencing the selected clone identified during the screening step. The sequence, including the N-terminal His6-tag (MRGSHHHHHHGS, SEQ ID NO: 3) and the C-terminal Cys-tag, was subcloned in Escherichia coli DH5a LacIq strain, following the procedure described in Huet S. et al, PLoS ONE 2015, 10(11): e014230410. Briefly, DNA amplicon encoding SEQ ID NO: 1 was subcloned in a plasmid derived from pQE-30 (Qiagen) by Gibson assembly to encode SEQ ID NO: 2, and the ligation mixture was transformed into Escherichia coli DH5a LacIq strains (Invitrogen). Clones were isolated and selected on 2×YT medium plates containing 100 μg/ml ampicillin and 25 μg/ml kanamycin. The plasmid construction was confirmed by Sanger sequencing. Precultures from transformed Escherichia coli DH5α LacIq were grown overnight at 37° C. in 2×YT medium with 1% glucose, 100 μg/ml ampicillin and 25 μg/ml kanamycin. Precultures were diluted 1:20 in 2×YT medium with 0.1% glucose, 100 μg/ml ampicillin and 25 μg/ml kanamycin, and grown at 37° C. to mid-log phase (OD600=0.8-1.0). Then, protein expression was induced by addition of Isopropyl β-D-1-thiogalactopyranoside to the final concentration of 0.5 mM and the culture shaken at 30° C. overnight. Bacteria were pelleted by 45 min centrifugation at 3220 g. Cell pellets were resuspended in a pH 7.4 lysis buffer composed of 1× BugBuster Protein Extraction Reagent, 5 μg/ml DNaseI, 20 mM Tris, 500 mM NaCl, and 25 mM Imidazole. Cell lysis occurred at room temperature for 1 h and the suspension was centrifuged at 3220 g for 45 min to remove cell debris. Histag-proteins were then purified from supernatants by immobilized metal ion affinity chromatography (IMAC), using His60 Nickel Superflow resin (Clontech) and a pH 7.4 elution buffer composed of 20 mM Tris, 500 mM NaCl, and 250 mM Imidazole. Additional endotoxin removal step was carried over for samples engaged in cell-based assay. First, samples were buffer-exchanged by dialysis against PBS (10 mM phosphate, 2.7 mM KCl and 137 mM NaCl, pH 7.4; Sigma-Aldrich). Then, samples were filtered on a Sartobind STIC PA anion exchanger (Sartorius). Finally, samples were dialyzed against PBS, filtered on Minisart hydrophilic membranes with 0.2 m pore size (Sartorius), then stored in sterile conditions.


Such derivatized affitin was named Aff02 (SEQ ID NO: 2).


Example 2: Determination of Cells Internalization of the Affitin Aff01

The cells internalization of Aff01 was evaluated using a dedicated cell-based assay. Briefly, Aff01 was tagged with SEQ ID NO: 3 (i.e. forming sequence Aff02-0) and then fused to a truncated portion of Pseudomonas exotoxin A, wherein the cell internalization domain of the toxin is removed, thus forming a final peptide with the sequence as set out in SEQ ID NO: 5. Such truncated form (Pe38) needs to be internalized by means of the Aff01 to keep its cytotoxic activity.


To test internalization, then, the viability of the breast adenocarcinoma cell line SK-BR-3 (overexpressing HER2 receptor) and of the breast cancer cell line MCF-7 (low expressing HER2 receptor) was measured at different nanofitin concentrations. A dose-dependent cytotoxic induction was observed on SK-BR-3 cells, while, on the contrary, no significant alterations were observed on MCF-7 cells. Cell death induction demonstrated an effective transport of Pe38 through HER2 receptor due to Aff01 triggering internalization.


Example 3: Coupling of Affitin Aff02 to IRDye800CW-Maleimide

IRDye800CW-maleimide was purchased (Li-Cor Inc., USA) in order to conjugate an activated form of this fluorophore IRDye800CW to the reactive cysteine-tag at the C-terminus of affitin Aff02 (SEQ ID NO: 2).


Since the terminal free cysteine is highly reactive, it also leeds to the formation of a dimeric form of the protein in stock solutions. A disulphide bridge between the Cys terminal residues of two monomers is indeed formed in the presence of oxygen. Therefore, a reduction step on the Aff02 stock solution prior to conjugation was performed. The Aff02 stock solution was reduced in phosphate buffer (0.02 M phosphate, 0.054 M KCl, 0.274 M NaCl, pH 7.4) by incubation with a mild reducing agent (TCEP). In details, 50 μL of 0.5 M of TCEP were added to 5 mg of affitin Aff02 and incubated for 1-16 h (preferably 16 h) at room temperature. After TCEP removal by size exclusion chromatography using a PD-10 desalting column, the IRDye800CW-maleimide (10 eq.) in DMSO was added for the coupling. After shaking overnight at room temperature, the crude product was purified by immobilized metal affinity chromatography (IMAC) on a Ni-NTA resin, exploiting the presence of the hexahistidine-tag on the affitin.


The fractions containing the conjugate IRDye800CW-Aff02 were collected and characterized by measuring their optical density from 230 to 830 nm by UV/VIS spectrophotometry, and by SE-HPLC (see FIG. 1) and RP-HPLC. IRDye800CW-Aff02 resulted at concentration of 2.34 mg/mL (Molar Extinction Coefficient: 12051.52 M−1 cm−1).


RP-HPLC analysis was also performed in order to check the reduction of the dimeric protein. The chromatograms of the dimer and of Aff02 conjugated to IRDye800CW showed that the reduction of disulfide bridges was efficiently performed prior to conjugation.


Example 4: In Vitro Affinity Assay of the Unlabeled Affitin Aff02-0 to HER2 Receptor

The affinity of the polypeptides of the invention for the target HER2 was measured by Biolayer interferometry assay. To this purpose, the protein A biosensors (ForteBio) were coated with human HER2 (hHER2) recombinant protein. After a brief wash with PBS, the coated sensor was assayed versus the affitin Aff02-0 at different concentrations (i.e. 10000, 500, 250, 125, 62.5, 31.3, 15.6 and 0 nM).


A curve fitting analysis was then performed in order to determine the dissociation equilibrium constant. The sensorgrams were analyzed using the Octet Data Analysis software 8.2 (ForteBio) with a 1:1 binding fit model to determine the dissociation equilibrium constant (Kd) and the related association and dissociation rate constants (kon and koff) with R2 value assessing the fitting quality.


The resulting kinetic profiles showed a Kd associated to Aff02-0 affitin in the nanomolar range, in particular a Kd of 1.8±0.1×10−8 M, as summarized in the following Table 1.









TABLE 1







Dissociation equilibrium constants of the hHER2-Aff02-0 complex









Unlabeled Aff02-0 affitin















Kd (M)
1.8 ± 0.1 × 10−8
M



kon (M−1s−1)
4.5 ± 0.1 × 105
M−1s−1



koff (s−1)
8.3 ± 0.1 × 10−3
s−1










R2
0.9626










Example 5: In Vitro Affinity Assay of the IRDye800CW-Aff02 Conjugate to HER2 Receptor

The affinity of the IRDye800CW-Aff02 conjugate to the target HER2 was measured by Biolayer interferometry assay. To this purpose, the protein A biosensors (ForteBio) were coated with human HER2 (hHER2) recombinant protein. After a brief wash with PBS, the coated sensor was assayed versus the IRDye800CW-Aff02 conjugate at different concentrations (i.e. 10000, 1000, 100, 10, 1, 0.1 ng/mL).


A steady state analysis was performed in order to determine the dissociation equilibrium constant. The sensorgrams were analyzed using the Octet Data Analysis software 8.2 (ForteBio) with a 1:1 binding fit model to determine the dissociation equilibrium constant (Kd). The resulting kinetic profiles showed a Kd associated to IRDye800CW-Aff02 conjugate in the nanomolar range, as reported in Table 2.


These data show that the coupling of the fluorophore to the C-terminal cysteine of affitin Aff02 did not significantly changed the Kd compared to the unlabeled Aff02-0.









TABLE 2







Dissociation equilibrium constant of the


complex IRDye800CW-Aff02 conjugate/hHER2









IRDye800CW-Aff02 conjugate














Kd (M)
0.16 ± 0.1 × 10−8 M










Example 6: Competitive ELISA of Aff02 Polypeptide Versus Trastuzumab or Pertuzumab

A competitive ELISA assay has been developed in order to investigate if the epitope of the HER2/Neu antigen recognized by Aff02 polypeptide was distinct from the epitope recognized by the humanized monoclonal antibodies trastuzumab and pertuzumab, actually used in therapeutic treatments of HER2-positive tumors.


The first experiment, aimed at evaluating the quality of the assay, was a competitive ELISA assay carried out between trastuzumab and trastuzumab-biotin. Briefly, a Medisorp transparent plastic plate (96 wells, Thermo Scientific), was coated (64 wells) with 2.5 μg/mL of HER2/Fc chimeric protein in TBS pH 7.4 (1 h, RT). The wells were blocked with 0.5% BSA in TBS (1 h, RT).


Then different concentrations of trastuzumab (1.6, 0.8, 0.4, 0.2, 0.1, 0.05, 0.025 and 0 μg/mL) were incubated together with 0.2 μg/mL of trastuzumab-biotin in each well in triplicate (1 h at RT) in TBS+0.1% Tween 20 (TBS-T). At the end of the incubation, the plate was washed three times with 250 μL of TBS-T and incubated with Streptavidin-HRP 1:10000 in TBS-T (1 h at RT). The plate was washed again 3 times with TBS-T and developed 5 min with TMB reagent (100 μL). The reaction was blocked with 50 μL of sulfuric acid 2 M and read at 450 nm.


The same experiment was then performed in three alternative assays with Aff02 vs trastuzumab-biotin or pertuzumab-biotin: Assay a) Different concentrations of Aff02 (10000, 1000, 100, 10, 1, 0.1 and 0.01 ng/mL) were incubated together with 0.2 μg/mL of trastuzumab-biotin or pertuzumab-biotin in each well in triplicate. As positive control, the same experiment was repeated using an antibody anti RSGH TAG-HRP (1:4000) in place of streptavidin-HRP, in order to evaluate the curve affinity of Aff02 to HER2 receptor. The ELISA results, plotted in the diagram of FIG. 2, suggested that the trastuzumab-biotin, pertuzumab-biotin and Aff02 are all binders for HER2 receptor but the epitopes recognized by trastuzumab and pertuzumab are different from that recognized by Aff02 affitin.


Assay b) Additional competitive ELISA experiment was performed using a variable trastuzumab-biotin and pertuzumab-biotin concentrations in the presence of fixed Aff02 concentration. Briefly, a Medisorp transparent plastic plate (96 wells, Thermo Scientific) was coated (64 wells) with 2.5 μg/mL of Her2/Fc chimeric protein in TBS pH 7.4 (1 h, RT). The wells was blocked with 0.5% BSA in TBS (1 h, RT). Then different concentrations of trastuzumab-biotin (20000, 2000, 200, 20, 2, 0.2, 0.02 and 0 ng/mL) were incubated together with 10 μg/mL of Aff02 in each well in duplicate (1 h at RT) in TBS+0.1% Tween 20 (TBS-T). At the end of the incubation, the plate was washed three times with 300 μL of TBS-T and incubated with Streptavidin-HRP 1:10000 or anti-RSGH TAG-HRP 1:4000 in TBS-T (1 h at RT). The plate was washed again 3 times with TBS-T and developed 5 min with TMB reagent (100 μL). The reaction was blocked with 50 μL of sulfuric acid 2 M and read at 450 nm. From the ELISA results it is clear that the curves of Aff02 detected with anti RSGH-Tag Ab have a negligible response decrease when the trastuzumab-biotin or pertuzumab-biotin were increased (see FIGS. 3a and 3b respectively).


Assay c) Another competitive ELISA assay was performed in the same conditions using a variable concentration of trastuzumab-biotin and pertuzumab-biotin against buffer (no competition) and Aff02 (competition) in the same experimental condition of last competitive ELISA.


Overall, the results showed no competition between the targeted molecules, as the curves of both trastuzumab and pertuzumab were equal in the presence or absence of Aff02 affitin (see FIGS. 4a and 4b respectively).


It is clear from the diagrams that Aff02 retained the binding properties to HER2 and that its specificity for the receptor was not affected by the presence of the two reference antibodies. In fact, the competition studies demonstrated that the epitope recognized by the Aff02 is separate and distinct from the epitopes recognized by trastuzumab and pertuzumab monoclonal antibodies, so they do not compete for binding to HER2.


Example 7: In Vivo Biodistribution of Conjugate IRDye800CW-Aff02

In vivo optical imaging biodistribution of IRDye800CW-Aff02 was evaluated in healthy mice. Optical Imaging experiments on three healthy mice were performed using an IVIS Spectrum system after administration of IRDye800CW-Aff02 at a dose of 10 nmol/mouse, in an administration volume of 0.2 mL. The experiments were carried out at 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 24 h and 48 hours after intravenous administration of the conjugate.


Regions of interest (ROIs) were drawn on a reference background healthy region (hind leg's muscle) of the mouse and on whole body for each fluorescence image at every time point to evaluate signal intensity in the tissues, expressed as Average Radiant Efficiency (see FIG. 5, reporting the fluorescence signal curves of IRDye800CW-Aff02 administered 10 nmol/mouse in Balb/c nu/nu mice).


Pharmacokinetic analysis of the signal was performed to evaluate the half time of the tested product by using a mono-exponential decay model and the results are reported in Table 3:









TABLE 3







Half life values for IRDye800CW-Aff02


in muscle and in the whole body










Muscle
Whole body















Half life (h)
2.122
2.196










The two computed half-life values thus obtained were consistent, giving an average value of 2.16 h. Such values of biological half-life make the Aff02 conjugate potentially suitable as alternative tool for use in fast imaging protocols, for instance in optical imaging.


After sacrifice, several organs were collected for the measure of residual fluorescence (after 48h). The measurements of fluorescence signal intensity have been obtained from the analysis of each organ selecting an area of interest (ROIs) at the center of each sample. The analyzed organs were: kidney, lung, spleen, liver, muscle and heart.



FIG. 6 shows the ex-vivo biodistribution of IRDye800CW-Aff02 in healthy mice at 48 h after injection at a dosage 10 nmol/mouse. The higher uptake of IRDye800CW-Aff02 in the kidneys shows that it is preferably cleared by the renal pathway instead of the hepatobiliary pathway.


REFERENCES



  • 1. Slamon D. J. et al., Science 1989, 244, 707-712

  • 2. McAfee J. G. et al, Biochemistry 1995, 34, 10063-10077

  • 3. Mouratou B. et al, PNAS 2007, 104(46), 17983-17988

  • 4. Goux M. et al., Bioconjugated Chem. 2017, 28, 2361-2371

  • 5. WO2009/080810 A1

  • 6. Orlova A. et al., Cancer Res. 2006, 66(8), 4339-4348

  • 7. WO2012/096760 A1

  • 8. WO2017/161096 A1

  • 9. Bornhorst J. A. et al, Methods Enzymol. 2000, 326, 245-254

  • 10. Huet S. et al, PLoS ONE 2015, 10(11): e0142304


Claims
  • 1. A polypeptide that specifically binds to Human Epidermal Growth Factor Receptor 2 (HER2), comprising the amino acid sequence set out in SEQ ID NO: 1.
  • 2. The polypeptide according to claim 1, consisting of the amino acid sequence set out in SEQ ID NO. 1.
  • 3. The polypeptide according to claim 1, comprising the amino acid sequence set out in SEQ ID NO: 2.
  • 4. The polypeptide according to claim 3, consisting of the amino acid sequence set out in SEQ ID NO: 2.
  • 5. The polypeptide according to claim 1, which is linked to an imaging agent.
  • 6. The polypeptide according to claim 5, wherein the imaging agent is selected from the group consisting of a fluorophore moiety, a magnetic or paramagnetic moiety, a radiolabeling isotope, an affinity label, an X-ray responsive moiety, an ultrasound responsive moiety, a photoacoustic responsive imaging moiety and a nanoparticle-based moiety.
  • 7. The polypeptide according to claim 6, wherein the fluorophore moiety is a cyanine dye.
  • 8. The polypeptide according to claim 6, wherein the radiolabeling isotope is selected from 18F, 124, 11C, 64Cu, 68Ga, 89Zr, 44Sc and 99mTc and other radioisotopes of indium, gallium, yttrium, bismuth, radioactinides and radiolanthanides.
  • 9. The polypeptide according to claim 6, wherein the affinity label is biotin.
  • 10. (canceled)
  • 11. (canceled)
  • 12. (canceled)
  • 13. A diagnostic or imaging composition comprising a polypeptide as defined in claim 5 with pharmaceutically acceptable carriers, excipients, diluents and/or additives.
  • 14. A method of imaging comprising: administering the composition according to claim 13 to a subject; andimaging the subject with a diagnostic device.
  • 15. The method according to claim 14, wherein said imaging is based on an imaging technique selected from magnetic resonance, positron-emission tomography (PET), computed tomography (CT), ultrasounds (US), photoacoustic imaging (PAI), near-infrared fluorescence (NIRF) and single photon emission computed tomography (SPECT), or correlated with optical imaging (OI).
  • 16. The polypeptide according to claim 2, which is linked to an imaging agent.
  • 17. The polypeptide according to claim 16, wherein the imaging agent is selected from the group consisting of a fluorophore moiety, a magnetic or paramagnetic moiety, a radiolabeling isotope, an affinity label, an X-ray responsive moiety, an ultrasound responsive moiety, a photoacoustic responsive imaging moiety and a nanoparticle-based moiety.
  • 18. The polypeptide according to claim 17, wherein the fluorophore moiety is a cyanine dye.
  • 19. The polypeptide according to claim 17, wherein the radiolabeling isotope is selected from 18F, 124I, 11C, 64Cu, 68Ga, 89Zr, 44Sc and 99mTc and other radioisotopes of indium, gallium, yttrium, bismuth, radioactinides and radiolanthanides.
  • 20. The polypeptide according to claim 17, wherein the affinity label is biotin.
  • 21. A diagnostic or imaging composition comprising a polypeptide as defined in claim 16 with pharmaceutically acceptable carriers, excipients, diluents and/or additives.
  • 22. A method of imaging comprising: administering the composition according to claim 21 to a subject; andimaging the subject with a diagnostic device.
  • 23. The method according to claim 22, wherein said imaging is based on an imaging technique selected from magnetic resonance, positron-emission tomography (PET), computed tomography (CT), ultrasounds (US), photoacoustic imaging (PAI), near-infrared fluorescence (NIRF) and single photon emission computed tomography (SPECT), or correlated with optical imaging (OI).
Priority Claims (1)
Number Date Country Kind
19217647.7 Dec 2019 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2020/086398 12/16/2020 WO