[go: up one dir, main page]

WO2025105948A1 - [99mtc]tc-ipd-l1 as a tetradecapeptide radiopharmaceutical for detecting and monitoring the overexpression of the pd-l1 protein - Google Patents

[99mtc]tc-ipd-l1 as a tetradecapeptide radiopharmaceutical for detecting and monitoring the overexpression of the pd-l1 protein Download PDF

Info

Publication number
WO2025105948A1
WO2025105948A1 PCT/MX2024/050024 MX2024050024W WO2025105948A1 WO 2025105948 A1 WO2025105948 A1 WO 2025105948A1 MX 2024050024 W MX2024050024 W MX 2024050024W WO 2025105948 A1 WO2025105948 A1 WO 2025105948A1
Authority
WO
WIPO (PCT)
Prior art keywords
ipd
hydrazine
protein
leu
radiopharmaceutical
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/MX2024/050024
Other languages
Spanish (es)
French (fr)
Inventor
Guillermina FERRO FLORES
Myrna Alejandra LUNA GUTIÉRREZ
Blanca Elí Ocampo García
Clara Leticia SANTOS CUEVAS
Nallely Patricia JIMÉNEZ MANCILLA
Erika Patricia AZORÍN VEGA
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Instituto Nacional de Investigaciones Nucleares
Original Assignee
Instituto Nacional de Investigaciones Nucleares
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Instituto Nacional de Investigaciones Nucleares filed Critical Instituto Nacional de Investigaciones Nucleares
Publication of WO2025105948A1 publication Critical patent/WO2025105948A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/534Production of labelled immunochemicals with radioactive label
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/60Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances involving radioactive labelled substances

Definitions

  • the present invention relates to a cyclic tetradecapeptide containing a hydrazine group on the lysine side chain for use as a novel inhibitor of the PD-L1 protein (PD-L1), which, upon labeling with 99mTc ( 99mTc - ⁇ PD-L1), functions as a novel radiopharmaceutical for imaging PD-L1 expression.
  • PD-L1 protein PD-L1 protein
  • the ⁇ PD-L1 ligand (-6.7 kcal/mol, AutoDock affinity) labeled with " mTc is obtained with a radiochemical purity >95%.
  • the radiopharmaceutical [ 99mTc ]Tc- ⁇ PD-L1 can detect PD-L1 positive tumor lesions ⁇ n vivo in patients undergoing whole-body scanning by single-photon emission computed tomography (SPECT) in molecular nuclear medicine.
  • SPECT single-photon emission computed tomography
  • Immune checkpoint inhibitor (ICI) therapy has emerged as an important therapeutic option for the treatment of various cancer types [1-5].
  • PD-1 programmed death receptor 1
  • PD-L1 has been shown to be a reproducible biomarker. It can be used to guide therapeutic decisions and monitor response [1].
  • PD-L1 expression is not only heterogeneous between and within tumor lesions, but is also highly dynamic, with accelerated changes in relatively short times. Therefore, the detection of PD-L1 by immunohistochemical techniques should be complemented by other diagnostic modalities.
  • SPECT single photon emission computed tomography
  • PET positron emission tomography
  • radiolabeled antibodies minibodies, aphibiods, and nanobodies
  • these molecules have a relatively high degree of immunogenicity and can even induce potentially adverse immunological effects, such as cytokine storms [6].
  • small peptide and non-peptide molecules accumulate more rapidly in tumor tissue, allowing for same-day imaging of patients [16-19].
  • the elimination of radiopharmaceuticals is rapid and occurs within a favorable timeframe of minutes to hours. They also have the advantage of being manufactured following Good Manufacturing Practices (GMP) protocols.
  • GMP Good Manufacturing Practices
  • small molecules have the advantage of high tissue penetration and cell membrane permeability to reach defined molecular targets [16-19].
  • several important peptide- and small-molecule-based inhibitors have been developed for PD-1/PD-L1 blockade [6,10,11,13,16-19], However, only one peptide has been used in humans for PET imaging [15] and none for SPECT imaging.
  • Our invention is based on the use of hydrazine (pyridine-hydrazine) outside of a peptide macrocycle, different from WL12 as it does not contain an -S- group in the macrocycle, the fundamental basis of the Bristol-Myers Squibb Company, US patent (Pat. No. 9,879,046).
  • Hydrazine as a side chain in the peptide macrocycle to be patented (PD-L1), generates a simultaneous double interaction (ionic interaction/salt bridge and hydrogen bond) between the carboxylate ion (-COO-) of Glu-58 and an ammonium ion (- NHs + ) of the hydrazine present in the PD-L1, where the hydrogen of the -NHs + forms strong hydrogen bonds with the oxygen of -COO.
  • the stabilization energy due to ionic interactions (salt bridge) is many times higher than that of hydrogen bonding interactions, which add up to a simultaneous interaction such as that generated in the PD-L1 molecule ( Figure 1). This interaction allowed obtaining a more rigid structure with anchoring and recognition.
  • a cyclic tetradecapeptide containing a hydrazine group on the lysine side chain is presented for use as a novel inhibitor of the PD-L1 protein (PD-L1), which, after labeling with mTc ( 99mTc - ⁇ PD-L1), functions as a novel radiopharmaceutical for imaging PD-L1 expression.
  • PD-L1 protein PD-L1 protein
  • mTc 99mTc - ⁇ PD-L1
  • the ⁇ PD-L1 ligand (-6.7 kcal/mol, AutoDock affinity) labeled with 99mTc is obtained with a radiochemical purity >90%.
  • the radiopharmaceutical [ 99mTc ]Tc- ⁇ PD-L1 can detect PD-L1-positive tumor lesions in vivo in patients undergoing whole-body scanning by single-photon emission computed tomography (SPECT) in molecular nuclear medicine.
  • SPECT single-photon emission computed tomography
  • Our invention is based on the use of the hydrazine group (-NH2-NH2) outside a peptide macrocycle, different from WL12 [cyclo(AcTyr-MeAla-Asn-Pro-His-Leu-Hyp-Trp- Ser-Trp(Me)-MeNle-MeNle-Orn-Cys)-Gly-NH2 (thioether bridge -S- between Cys 14 and Ac-Tyr)] by not containing a -S- group within the peptide sequence of the macrocycle, the fundamental basis of the Bristol-Myers Squibb Company, US patent (Pat. No. 9,879,046).
  • hydrazine-nicotinoyl-PD-L1 [cyclo(Trp-Ser-Trp-Leu-Leu-Lys(hydrazine-nicotinoyl)-Cys-Tyr-Ala-Asn-Pro-H ⁇ s-Leu-Pro)] (PD-L1 ), generates a simultaneous double interaction (ionic interaction/salt bridge and hydrogen bond) between the carboxylate ion (-COO-) of Glu-58 and an ammonium ion (-NHs + ) of the hydrazine present in the ⁇ PD-L1 , where the hydrogen of the -NHs + forms strong hydrogen bonds with the oxygen of -COO .
  • a patient with advanced plantar malignant melanoma received [ 99m Tc]Tc- ⁇ PD-L1 and underwent a whole-body scan to differentiate between PD-L1-positive (with [ 99m Tc]Tc- ⁇ PD- L1 uptake) tumor lesions (visualized by their high metabolic activity with [ 18 F]FDG) and PD-L1-negative (without [ 99m Tc]Tc- ⁇ PD-L1 uptake) tumor lesions.
  • Molecular docking (Docking)
  • the three-dimensional structure of each ligand was exported in mol2 format using Chem3D to optimize the molecular geometry using the MMFF94 force field via the OpenBabel chemistry toolbox.
  • Human programmed death-1 protein was used as the receptor and its ligand (PD-L1 ) obtained from the protein database (PDB ID: 4ZQK), which was edited to remove water molecules, ions, and structures unrelated to PD-L1 in the X-ray crystallography process.
  • PDB ID: 4ZQK protein database
  • the receptor as a macromolecule and the corresponding ligands were prepared using the graphical package AutoDock Tools 1.5.7, with the search box cubically fixed 80 ⁇ on the x, y, and z axes, centered on the macromolecule. Semi-rigid docking was performed with AutoDock vina 1.1.2 around the entire receptor surface, generating 20 poses for each ligand. Finally, the inhibition constants were generated (Table 1).
  • Succinimidyl 6-Boc-hydrazinopyridine-3-carboxylate (NHS-HYNIC-Boc) was purchased from Synchem UG & Co (Felsberg, Germany). Cyclo(Trp-Ser-Trp-Leu-Leu-Lys-Cys-Tyr-Ala- Asn-Pro-His-Leu-Pro) (unconjugated PD-L1) was ordered to be custom synthesized according to our design and exclusively for our laboratory by Shanghai Yaxian Chemical Co, Ltd (Jiading, Shanghai, China). All other reagents were purchased from Merck (Burlington, MA, USA).
  • the hydrazine-nicotinoyl-iPD-L1 peptide ( Figure 2) (6 mg; 3.5 pmol) was dissolved in 0.5 mL of dimethylformamide (DMF), followed by the addition of N,N-diisopropylethylamine (20 pL).
  • NHS-HYNIC-Boc (2 mg; 5.7 pmol) was dissolved in 100 ⁇ l of DMF and added to the peptide solution for 24 h at room temperature. Unreacted NHS-HYNIC-Boc was removed from the reaction mixture by dialysis (Tube-O-DIALYZERTM mini dialysis system, 1 kDa MWCO; Merck; Burlington, MA, USA) for 24 h using injectable water.
  • the conjugate was purified by HPLC (Discovery® C18 HPLC column, 5 pm particle size, L x Dl 25 cm x 10 mm) (Merck; Burlington, MA, USA) using a linear gradient of 0.1% TFA-water/0.1% TFA-acetonitrile from 100 to 20% of the aqueous phase in 20 min at a flow rate of 4 mL/min. The fraction collected between 13.5 and 15 min was lyophilized.
  • the resulting powder was analyzed by FT-IR vibrational spectroscopy (400-4000 cm-1 , 50 scans at 0.4 cm-1 ; FT-IR 660 spectrometer, Agilent Technologies).
  • the peptide hydrazine-nicotinoyl-iPD-L1 dissolved in 40:60 ethane-water was used for characterization by UPLC mass spectroscopy (ADQUITY UPLC H-Class with QDa mass detector; Waters Corporation, Milford, USA) and UV-vis spectroscopy in the range of 200-400 nm (PerkinElmer LambdaBio spectrometer; Waltham, Massachusetts, USA).
  • the IR and mass spectra showed characteristic vibrational bands and molecular masses corresponding to the peptide hydrazine-nicotinoyl-iPD-L1 ( Figure 3).
  • Radiolabeled radiolabeled
  • the hydrazine-nicotinoyl-iPD-L1 peptide was dissolved in a 1:1 ethanol:water solution at a concentration of 1 mg/mL.
  • m Tc formation of the complex [ 99m Tc]Tc- ⁇ PD-L1
  • 100 pL of peptide solution, 500 pL of ethylenediamine-N,N'-diacetic acid (EDDA)/tricine solution (30 mg of EDDA in 1 .5 mL of 0.1 M NaOH / 60 mg of tricine in 1 .5 mL of 0.2 M phosphate buffer at a concentration of 1 mg / mL.
  • Sodium dodecyl sulfate-polyacrylamide gel electrophoresis was performed. The distribution of radioactivity was detected using a radio-TLC scanner (Mini-Gita, 60-150 keV, BGO-Crystal, window 25 x 2 mm, thickness 5 mm, resolution optimized for Tc-99m; RayTest, Kunststoff, Germany) to detect the binding of [ 99m Tc]Tc-IPD-L1 to the PD-L1 protein.
  • HCC827, HCT116, and C6 cell lines were fixed with 4% paraformaldehyde for 20 min, permeabilized with 0.5% TritonX-100, and blocked with 1% bovine serum albumin. Cells were then incubated overnight with an anti-PD-L1 antibody according to the supplier's instructions (FineTest, Wuhan Fine Biotech Co., China). Cells were then incubated with Alexa Fluor 488-conjugated goat anti-rabbit IgG (H+L) (Invitrogen Cat. No. A32731) for 1 h. DAPI was used to observe the fluorescence intensity in cells for nuclear staining. Fluorescence was observed by microscopy (Meiji Techno; model MT6200; Saitama, Japan).
  • the liquid was removed, and the button activity, corresponding to the percentage of cellular uptake (surface uptake) with respect to the initial activity, was measured.
  • a mixture of acetic acid/0.5 M NaCl was added, and centrifugation was repeated, followed by activity measurement.
  • the liquid was removed, and the button activity, corresponding to the percentage of activity internalized by the cells with respect to the uptake activity (fraction of internalization of surface uptake), was measured.
  • the percentage of cellular uptake of [ 99m Tc]Tc- ⁇ PD-L1 correlated with the expression of Cellular PD-L1 was determined by immunofluorescence in each of the cell lines analyzed. Uptake was zero for 99m TcO4Na (negative control) in all cells analyzed.
  • mice Male Nu/Nu mice (CINVESTAV, I.PN., Mexico City), 6-8 weeks old, were maintained in an aseptic barrier environment. Mice were inoculated by subcutaneous injection of 1 x106 HCC827 cells/0.1 mL PBS into the upper back. Injection sites were periodically monitored for tumor progression. On day 10 after tumor cell inoculation, animals were injected into the tail vein with 18.5 MBq (50 pL) of [ 99m Tc]Tc- ⁇ PD-L1.
  • [ 99mTc ]Tc- ⁇ PD-L1 has the ability to significantly uptake and detect with high sensitivity tumors and metastases with PD-L1 expression, due to a simultaneous double interaction (ionic/salt bridge interaction and hydrogen bonding) between the carboxylate ion (-COO-) of Glu-58 and an ammonium ion (-NHs + ) of the hydrazine present in ⁇ PD-L1 , where the hydrogen of -NHs + forms strong hydrogen bonds with the oxygen of -COO .
  • the stabilization energy due to ionic interactions (salt bridge) is many times higher than that of hydrogen bonding interactions, which add up to a simultaneous interaction such as that generated in the ⁇ PD-L1 molecule.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Analytical Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Radiology & Medical Imaging (AREA)
  • Optics & Photonics (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to a cyclic tetradecapeptide containing a hydrazine group in the side chain of the lysine for use as a novel PD-L1 protein inhibitor (iPD-L1). The hydrazine as a side chain in the macrocycle of the tetradecapeptide to be patented, referred to as iPD-L1 [cyclo(Trp-Ser-Trp-Leu-Leu-Lys(hydrazine-nicotinoyl)-Cys-Tyr-Ala-Asn-Pro-His-Leu-Pro)], generates a simultaneous dual interaction (ionic/salt bridge interaction and hydrogen bond) between the carboxylate ion (-COO-) of the Glu-58 and an ammonium ion (-NH3 +) of the hydrazine present in the iPD-L1, the hydrogen of the - NH3 + forming strong hydrogen bonds with the oxygen of -COO-. Said interaction allows a rigid structure to be obtained with sufficient anchorage to and recognition of the PD-L1 ligand. The iPD-L1 ligand (-6.7 kcal/mol, AutoDock affinity) labelled with 99mTc ([99mTc]Tc-iPD-L1) is obtained with >95% radiochemical purity and is capable of detecting the PD-L1 protein in vivo by means of single photon emission computerised tomography (SPECT). The purpose of this invention is to provide a new specific SPECT radiopharmaceutical (molecular target radiopharmaceutical) with high sensitivity for the detection of tumours with PD-L1 protein overexpression.

Description

[99mTc]Tc-iPD-L1 como un radiofármaco tetradecapeptídico para la detección y seguimiento de la sobreexpresión de la proteína PD-L1 [ 99m Tc]Tc-iPD-L1 as a tetradecapeptide radiopharmaceutical for the detection and monitoring of PD-L1 protein overexpression

DESCRIPCIÓN DESCRIPTION

CAMPO TÉCNICO DE LA INVENCIÓN TECHNICAL FIELD OF THE INVENTION

La presente invención se refiere a un tetradecapéptido cíclico conteniendo un grupo hidracina en la cadena lateral de la lisina para su uso como un nuevo inhibidor de la proteína PD-L1 (¡PD-L1 ), el cual, tras su marcado con 99mTc (99mTc-¡PD-L1 ), funciona como un nuevo radiofármaco para la obtención de imágenes de la expresión de PD-L1 . El ligando ¡PD-L1 (-6.7 kcal/mol, afinidad AutoDock) marcado con "mTc se obtiene con una pureza radioquímica >95%. El radiofármaco [99mTc]Tc-¡PD-L1 puede detectar lesiones tumorales positivas a PD-L1 ¡n vivo en pacientes sometidos a un escaneo de cuerpo entero por tomografía computarizada de emisión de fotón único (SPECT) en medicina nuclear molecular. The present invention relates to a cyclic tetradecapeptide containing a hydrazine group on the lysine side chain for use as a novel inhibitor of the PD-L1 protein (PD-L1), which, upon labeling with 99mTc ( 99mTc -¡PD-L1), functions as a novel radiopharmaceutical for imaging PD-L1 expression. The ¡PD-L1 ligand (-6.7 kcal/mol, AutoDock affinity) labeled with " mTc is obtained with a radiochemical purity >95%. The radiopharmaceutical [ 99mTc ]Tc-¡PD-L1 can detect PD-L1 positive tumor lesions ¡n vivo in patients undergoing whole-body scanning by single-photon emission computed tomography (SPECT) in molecular nuclear medicine.

ANTECEDENTES BACKGROUND

La terapia con inhibidores de puntos de control inmunitarios ( ICI) se ha convertido en una importante opción terapéutica para el tratamiento de diversos tipos de cáncer [1 -5]. Entre los puntos de control inmunitarios, el receptor de muerte programada 1 (PD-1) y su ligando (PD-L1 ) son las dianas moleculares clave para el tratamiento con ICI. PD-L1 ha demostrado ser un biomarcador reproducible. Puede utilizarse para guiar las decisiones terapéuticas y monitorizar la respuesta [1 ]. Sin embargo, la expresión de PD- L1 no sólo es heterogénea entre las lesiones tumorales y dentro de ellas, sino que también es muy dinámica y con cambios acelerados en tiempos relativamente cortos. Por lo tanto, la detección de PD-L1 mediante técnicas inmunohistoquímicas debe complementarse con otras modalidades de diagnóstico. Por otro lado, las técnicas de imagen molecular como la tomografía por emisión de fotón único (SPECT) y la tomografía por emisión de positrones (PET) tienen la ventaja de obtener imágenes ¡n vivo no invasivas de cuerpo entero, con alta sensibilidad, adecuada resolución espacial y tiempos de adquisición de imagen en minutos. Por lo tanto, puede visualizarse la expresión heterogénea del gen PD-L1 entre todas las lesiones tumorales de los pacientes a lo largo del tiempo [6-14], El desarrollo preclínico de radiofármacos inhibidores de PD-L1 para SPECT y PET se basa en tres tipos diferentes de moléculas: anticuerpos, péptidos y pequeñas moléculas no peptídicas [6]. A nivel preclínico y clínico, se ha observado una elevada acumulación tisular de diversos tipos de anticuerpos radiomarcados (minicuerpos, afibiodes y nanocuerpos) [7,8,12-14], Asimismo, estas moléculas tienen un grado relativamente alto de inmunogenicidad e incluso pueden inducir efectos inmunológicos potencialmente adversos, como las tormentas de citoquinas [6], En cambio, las pequeñas moléculas peptídicas y no peptídicas se acumulan más rápidamente en el tejido tumoral, lo que permite obtener imágenes de los pacientes en el mismo día [16-19], Asimismo, la eliminación de los radiofármacos es rápida y se produce en un plazo favorable de minutos a horas. También tienen la ventaja de que se fabrican con protocoles de Buenas Prácticas de Fabricación (BPF). Además, las moléculas pequeñas tienen la ventaja de su alta penetración tisular y permeabilidad de la membrana celular para alcanzar dianas moleculares definidas [16-19], En los últimos años, se han desarrollado varios inhibidores importantes basados en péptidos y moléculas pequeñas para el bloqueo de PD-1/PD-L1 [6,10,11 ,13,16-19], No obstante, sólo un péptido se ha utilizado en humanos para la obtención de imágenes PET [15] y ninguno para la obtención de imágenes SPECT.Immune checkpoint inhibitor (ICI) therapy has emerged as an important therapeutic option for the treatment of various cancer types [1-5]. Among immune checkpoints, programmed death receptor 1 (PD-1) and its ligand (PD-L1) are the key molecular targets for ICI treatment. PD-L1 has been shown to be a reproducible biomarker. It can be used to guide therapeutic decisions and monitor response [1]. However, PD-L1 expression is not only heterogeneous between and within tumor lesions, but is also highly dynamic, with accelerated changes in relatively short times. Therefore, the detection of PD-L1 by immunohistochemical techniques should be complemented by other diagnostic modalities. On the other hand, molecular imaging techniques such as single photon emission computed tomography (SPECT) and positron emission tomography (PET) have the advantage of obtaining noninvasive, in vivo images of the whole body, with high sensitivity, adequate spatial resolution and image acquisition times in minutes. Therefore, the heterogeneous expression of the PD-L1 gene among all tumor lesions of patients over time can be visualized [6-14]. The preclinical development of PD-L1 inhibitory radiopharmaceuticals for SPECT and PET is based on three different types of molecules: antibodies, peptides, and small non-peptide molecules [6]. At preclinical and clinical levels, a high tissue accumulation of various types of radiolabeled antibodies (minibodies, aphibiods, and nanobodies) has been observed [7,8,12-14]. Furthermore, these molecules have a relatively high degree of immunogenicity and can even induce potentially adverse immunological effects, such as cytokine storms [6]. In contrast, small peptide and non-peptide molecules accumulate more rapidly in tumor tissue, allowing for same-day imaging of patients [16-19]. Furthermore, the elimination of radiopharmaceuticals is rapid and occurs within a favorable timeframe of minutes to hours. They also have the advantage of being manufactured following Good Manufacturing Practices (GMP) protocols. Furthermore, small molecules have the advantage of high tissue penetration and cell membrane permeability to reach defined molecular targets [16-19], In recent years, several important peptide- and small-molecule-based inhibitors have been developed for PD-1/PD-L1 blockade [6,10,11,13,16-19], However, only one peptide has been used in humans for PET imaging [15] and none for SPECT imaging.

En 2022 se publicó la primera imagen PET humana de la expresión de PD-L1 en pacientes con cáncer de pulmón, con una excelente captación y especificidad [15], El péptido utilizado fue un ciclopéptido (14 aminoácidos) denominado WL12 (péptido conteniendo un tiol dentro de un macrociclo y patentado por Bristol-Myers Squibb Company, U.S. Pat. No. 9,879,046) [9], con cuatro aminoácidos mediados [Trp(Me), NMeAla y dos NMeNle] y una cadena lateral del residuo Orn conjugada al 68Ga-NOTA para la obtención de imágenes PET. In 2022, the first human PET image of PD-L1 expression in lung cancer patients was published, with excellent uptake and specificity [15], The peptide used was a cyclopeptide (14 amino acids) called WL12 (peptide containing a thiol within a macrocycle and patented by Bristol-Myers Squibb Company, US Pat. No. 9,879,046) [9], with four mediated amino acids [Trp(Me), NMeAla and two NMeNle] and a side chain of the Orn residue conjugated to 68 Ga-NOTA for PET imaging.

Nuestra invención se basa en el uso de la hidracina (piridina-hidracina) fuera de un macrociclo peptídico, diferente al WL12 al no contener un grupo -S- en el macrociclo, base fundamental de la patente Bristol-Myers Squibb Company, U.S. (Pat. No. 9,879,046). La hidracina como cadena lateral en el macrociclo del péptido a patentar (¡PD-L1 ), genera una doble interacción simultánea, (interacción iónica/puente salino y enlace de hidrógeno) entre el ion carboxilato (-COO-) de la Glu-58 y un ion amonio (- NHs+) de la hidracina presente en el ¡PD-L1 , donde el hidrógeno del -NHs+ forma fuertes enlaces de hidrógeno con el oxígeno de -COO . Es de conocimiento general, que la energía de estabilización debida a las interacciones iónicas (puente salino) es muchas veces superior a la de las interacciones de enlace de hidrógeno, las cuales se suman en una interacción simultánea como la generada en la molécula de ¡PD-L1 (Figura 1 ). Dicha interacción permitió obtener una estructura más rígida con anclaje y reconocimiento suficiente al ligando PD-L1 sin tener que utilizar, en el nuevo macrociclo peptídico ¡PD- L1 a patentar, sitios de metilación en los aminoácidos, ni enlaces -S- como en la cadena cíclica, como ocurre en el WL12. Además, Orn se sustituyó por Lys para obtener una cadena lateral más larga que no interfiriera con el sitio de reconocimiento, mejorando así la afinidad del péptido inhibidor de PD-L1 (¡PD-L1 ) (Tabla 1). Aún tras la conjugación 99mTc-HYNIC (imagen SPECT), la ventaja de la hidracina es su doble grupo amino, por lo que la interacción del amino no terminal con Glu-58(PD-L1 ) existe dada la cercanía con Glu-58 (4.88 nm) (Figura 1 ). Our invention is based on the use of hydrazine (pyridine-hydrazine) outside of a peptide macrocycle, different from WL12 as it does not contain an -S- group in the macrocycle, the fundamental basis of the Bristol-Myers Squibb Company, US patent (Pat. No. 9,879,046). Hydrazine as a side chain in the peptide macrocycle to be patented (PD-L1), generates a simultaneous double interaction (ionic interaction/salt bridge and hydrogen bond) between the carboxylate ion (-COO-) of Glu-58 and an ammonium ion (- NHs + ) of the hydrazine present in the PD-L1, where the hydrogen of the -NHs + forms strong hydrogen bonds with the oxygen of -COO. It is common knowledge that the stabilization energy due to ionic interactions (salt bridge) is many times higher than that of hydrogen bonding interactions, which add up to a simultaneous interaction such as that generated in the PD-L1 molecule (Figure 1). This interaction allowed obtaining a more rigid structure with anchoring and recognition. sufficient to the PD-L1 ligand without having to use, in the new peptide macrocycle ¡PD- L1 to be patented, methylation sites in the amino acids, or -S- bonds as in the cyclic chain, as occurs in WL12. In addition, Orn was replaced by Lys to obtain a longer side chain that would not interfere with the recognition site, thus improving the affinity of the PD-L1 inhibitory peptide ¡PD-L1 ) (Table 1). Even after 99m Tc-HYNIC conjugation (SPECT image), the advantage of hydrazine is its double amino group, so the interaction of the non-terminal amino with Glu-58 (PD-L1 ) exists given the proximity to Glu-58 (4.88 nm) (Figure 1 ).

Tabla 1. Afinidad y constante de inhibición de derivados peptídicos macrocíclicos determinados por docking molecular (AutoDock). Table 1. Affinity and inhibition constant of macrocyclic peptide derivatives determined by molecular docking (AutoDock).

Péptido Afinidad Constante Sitio de unión inhibidor (kcal/mol) de inhibición Peptide Inhibitory Binding Site Affinity Constant (kcal/mol)

(pM) (pM)

Lys25, Lys41 , Leu94, Leu27, ¡PD-L1 -Lys25, Lys41, Leu94, Leu27, ¡PD-L1 -

Phe42, Val23. Glu39, Lys124, macrociclo no -6.7 12.26 Phe42, Val23. Glu39, Lys124, macrocycle no -6.7 12.26

Pro24, Thr22, Val44 conjugado Pro24, Thr22, Val44 conjugate

Gln107, Arg125, Alai 32, hydracina- Leu106, Valí 11 , lie 126, Glu60, nicotinoil-iPD- -7.2 5.27 Alai 09, Met5, Thr127, Arg1 13, Gln107, Arg125, Alai 32, hydrazine- Leu106, Valí 11, lie 126, Glu60, nicotinoyl-iPD- -7.2 5.27 Alai 09, Met5, Thr127, Arg1 13,

L1 (¡PD-L1 ) Lys129, Tyr1 12 L1 (PD-L1 ) Lys129, Tyr1 12

WL12- -5.1 82.51 Ser34, Glu39, Glu31 , Asn35 macrociclo no conjugado hydracina-WL12- -5.1 82.51 Ser34, Glu39, Glu31, Asn35 non-conjugated macrocycle hydrazine-

Leu94, Leu88, Glu31 , Gln100, nicotinoil - -6.0 39.95 Leu94, Leu88, Glu31, Gln100, nicotinoyl - -6.0 39.95

Thr102, Ser34 Thr102, Ser34

WL12 Referencias WL12 References

[1 ]. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Sosman, J. A.; Atkins, M.B.; Leming, P.D. Five-year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non-small cell lung cancer treated with nivolumab. JAMA oncology 2019, 5, 1411 - 1420. [1]. Topalian, SL; Hodi, F.S.; Brahmer, JR; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Sosman, JA; Atkins, M.B.; Leming, PD Five-year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non-small cell lung cancer treated with nivolumab. JAMA oncology 2019, 5, 1411 - 1420.

[2], Cho, D.C.; Sosman, J.A.; Sznol, M.; Gordon, M.S.; Hollebecque, A.; Hamid, O.; [2], Cho, D.C.; Sosman, J.A.; Sznol, M.; Gordon, M.S.; Hollebecque, A.; Hamid, O.;

McDermott, D.F.; Delord, J. -P.; Rhee, I.P.; Mokatrin, A. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2013, 31 (15_suppl) 4505-4505. McDermott, D.F.; Delord, J.-P.; Rhee, I.P.; Mokatrin, A. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (mRCC). Journal of Clinical Oncology 2013, 31 (15_suppl) 4505-4505.

[3], Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.-J.; Kefford, R.; Wolchok, J.D.; [3], Hamid, O.; Robert, C.; Daud, A.; Hodi, F.S.; Hwu, W.-J.; Kefford, R.; Wolchok, J.D.;

Hersey, P.; Joseph, R.W.; Weber, J.S. Safety and tumor responses with lambrolizumab (anti-PD-1 ) in melanoma. New England Journal of Medicine 2013, 369, 134-144. Hersey, P.; Joseph, R.W.; Weber, J.S. Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. New England Journal of Medicine 2013, 369, 134-144.

[4], Topalian, S.L.; Sznol, M.; McDermott, D.F.; Kluger, H.M.; Carvajal, R.D.; Sharfman,[4], Topalian, S.L.; Sznol, M.; McDermott, D.F.; Kluger, H.M.; Carvajal, R.D.; Sharfman,

W.H.; Brahmer, J.R.; Lawrence, D.P.; Atkins, M.B.; Powderly, J.D. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. Journal of Clinical Oncology 2014, 32, 1020. W.H.; Brahmer, J.R.; Lawrence, D.P.; Atkins, M.B.; Powderly, J.D. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. Journal of Clinical Oncology 2014, 32, 1020.

[5]. Weber, J.; Minor, D.; D'Angelo, S.; Hodi, F.; Gutzmer, R.; Neyns, B.; Hoeller, C.; [5]. Weber, J.; Minor, D.; D'Angelo, S.; Hodi, F.; Gutzmer, R.; Neyns, B.; Hoeller, C.;

Khushalani, N.; Miller, W.; Grob, J. A phase 3 randomized, open-label study of nivolumab (anti-PD-1 ; BMS-936558; ONO-4538) versus investigator's choice chemotherapy (ICC) in patients with advanced melanoma after prior anti-CTLA-4 therapy. Annals of Oncology 2014, 25, v1 . Khushalani, N.; Miller, W.; Grob, J. A phase 3 randomized, open-label study of nivolumab (anti-PD-1 ; BMS-936558; ONO-4538) versus investigator's choice chemotherapy (ICC) in patients with advanced melanoma after prior anti-CTLA-4 therapy. Annals of Oncology 2014, 25, v1.

[6]. Krutzek, F.; Kopka, K.; Stadlbauer, S. Development of Radiotracers for Imaging of the PD-1/PD-L1 Axis. Pharmaceuticals 2022, 15, 747. [6]. Krutzek, F.; Kopka, K.; Stadlbauer, S. Development of Radiotracers for Imaging of the PD-1/PD-L1 Axis. Pharmaceuticals 2022, 15, 747.

[7], Liu T, Wu Y, Shi L, Li L, Hu B, Wang Y, Gao H, Yu X, Zhang X, Zhao H, Wan Y,[7], Liu T, Wu Y, Shi L, Li L, Hu B, Wang Y, Gao H, Yu X, Zhang X, Zhao H, Wan Y,

Jia B, Wang F. Preclinical evaluation of [99mTc]Tc-labeled anti-EpCAM nanobody for EpCAM receptor expression imaging by immuno-SPECT/CT. Eur J Nucl Med Mol Imaging. 2022 49(6) :1810-1821 . Jia B, Wang F. Preclinical evaluation of [99mTc]Tc-labeled anti-EpCAM nanobody for EpCAM receptor expression imaging by immuno-SPECT/CT. Eur J Nucl Med Mol Imaging. 2022 49(6) :1810-1821.

[8], Zhang, Y.; Ding, Y.; Li, N.; Wang, S.; Zhou, S.; Li, R.; Yang, H.; Li, W.; Qu, J. [8], Zhang, Y.; Ding, Y.; Li, N.; Wang, S.; Zhou, S.; Li, R.; Yang, H.; Li, W.; What, J.

Noninvasive Imaging of Tumor PD-L1 Expression Using [99mTc]Tc-Labeled KN035 with SPECT/CT. Molecular Pharmaceutics 2022, 20, 690-700. Noninvasive Imaging of Tumor PD-L1 Expression Using [99mTc]Tc-Labeled KN035 with SPECT/CT. Molecular Pharmaceutics 2022, 20, 690-700.

[9]. Miller MM, et al. Macrocyclic inhibitors of the PD-1/PD-L1 and CD80(B7-1 )/PD-L1 protein/protein interactions, US patent number: 9879046, Type: Grant Filed: July 7, 2016 Date of Patent: January 30, 2018, Assignee: Bristol-Myers Squibb Company. [9]. Miller MM, et al. Macrocyclic inhibitors of the PD-1/PD-L1 and CD80(B7-1 )/PD-L1 protein/protein interactions, US patent number: 9879046, Type: Grant Filed: July 7, 2016 Date of Patent: January 30, 2018, Assignee: Bristol-Myers Squibb Company.

[10]. Chatterjee S, Lesniak WG, Miller MS, Lisok A, Sikorska E, Wharram B, Kumar D,[10]. Chatterjee S, Lesniak WG, Miller MS, Lisok A, Sikorska E, Wharram B, Kumar D,

Gabrielson M, Pomper MG, Gabelli SB, Nimmagadda S. Rapid PD-L1 detection in tumors with PET using a highly specific peptide. Biochem Biophys Res Commun. 2017 Jan 29;483(1 ):258-263. Gabrielson M, Pomper MG, Gabelli SB, Nimmagadda S. Rapid PD-L1 detection in tumors with PET using a highly specific peptide. Biochem Biophys Res Commun. 2017 Jan 29;483(1):258-263.

[11 ]. De Silva, R.A.; Kumar, D.; Lisok, A.; Chatterjee, S.; Wharram, B.; Venkateswara[11]. De Silva, R.A.; Kumar, D.; Lisok, A.; Chatterjee, S.; Wharram, B.; Venkateswara

Rao, K.; Mease, R.; Dannals, R.F.; Pomper, M.G.; Nimmagadda, S. Peptide-based 68Ga-PET radiotracer for imaging PD-L1 expression in cancer. Molecular Pharmaceutics 2018, 15, 3946-3952. Rao, K.; Mease, R.; Dannals, R.F.; Pomper, M.G.; Nimmagadda, S. Peptide-based 68Ga-PET radiotracer for imaging PD-L1 expression in cancer. Molecular Pharmaceutics 2018, 15, 3946-3952.

[12], Hughes, D.J.; Chand, G.; Johnson, J.; Bailey, D.; Adamson, K.; Goh, V.; Cook,[12], Hughes, D.J.; Chand, G.; Johnson, J.; Bailey, D.; Adamson, K.; Goh, V.; Cook,

G.J. Inter-rater and intra-rater agreement of [99mTc]-labelled NM-01 , a singledomain programmed death-ligand 1 (PD-L1 ) antibody, using quantitative SPECT/CT in non-small cell lung cancer. EJNMMI research 2023, 13, 51 G.J. Inter-rater and intra-rater agreement of [99mTc]-labelled NM-01, a singledomain programmed death-ligand 1 (PD-L1) antibody, using quantitative SPECT/CT in non-small cell lung cancer. EJNMMI research 2023, 13, 51

[13]. Li, W.; Wang, Y.; Rubins, D.; Bennacef, I.; Holahan, M.; Haley, H.; Purcell, M.; [13]. Li, W.; Wang, Y.; Rubins, D.; Bennacef, I.; Holahan, M.; Haley, H.; Purcell, M.;

Gantert, L.; Hseih, S.; Judo, M. PET/CT imaging of 89 Zr-N-sucDf-Pembrolizumab in healthy cynomolgus monkeys. Molecular Imaging and Biology 2021 , 23, 250- 259. Gantert, L.; Hseih, S.; Judo, M. PET/CT imaging of 89 Zr-N-sucDf-Pembrolizumab in healthy cynomolgus monkeys. Molecular Imaging and Biology 2021, 23, 250-259.

[14], Hughes, D.J.; Chand, G.; Goh, V.; Cook, G.J. Inter-and intraobserver agreement of the quantitative assessment of [99mTc]-labelled anti-programmed death-ligand 1 (PD-L1 ) SPECT/CT in non-small cell lung cancer. EJNMMI research 2020, 10 (1 ):145. [15]. Zhou, X.; Jiang, J.; Yang, X.; Liu, T.; Ding, J.; Nimmagadda, S.; Pomper, M.G.;[14], Hughes, D.J.; Chand, G.; Goh, V.; Cook, GJ Inter-and intraobserver agreement of the quantitative assessment of [99mTc]-labeled anti-programmed death-ligand 1 (PD-L1 ) SPECT/CT in non-small cell lung cancer. EJNMMI research 2020, 10 (1):145. [15]. Zhou, X.; Jiang, J.; Yang, X.; Liu, T.; Ding, J.; Nimmagadda, S.; Pomper, M.G.;

Zhu, H.; Zhao, J.; Yang, Z. First-in-Humans evaluation of a PD-L1-binding peptide PET radiotracer in non-small cell lung cancer patients. Journal of Nuclear Medicine 2022, 63, 536-542. Zhu, H.; Zhao, J.; Yang, Z. First-in-Humans evaluation of a PD-L1-binding peptide PET radiotracer in non-small cell lung cancer patients. Journal of Nuclear Medicine 2022, 63, 536-542.

[16]. Guzik, K.; Tómala, M.; Muszak, D.; Konieczny, M.; Hec, A.; Blaszkiewicz, U.; [16]. Guzik, K.; Take it, M.; Muszak, D.; Konieczny, M.; Hec, A.; Blaszkiewicz, U.;

Pústula, M.; Butera, R.; Domling, A.; Holak, T.A. Development of the inhibitors that target the PD-1/PD-L1 interaction — a brief look at progress on small molecules, peptides and macrocycles. Molecules 2019, 24, 2071 . Pustule, M.; Butera, R.; Domling, A.; Holak, T.A. Development of the inhibitors that target the PD-1/PD-L1 interaction — a brief look at progress on small molecules, peptides and macrocycles. Molecules 2019, 24, 2071.

[17], Chen, T.; Li, Q.; Liu, Z.; Chen, Y.; Feng, F.; Sun, H. Peptide-based and small synthetic molecule inhibitors on PD-1/PD-L1 pathway: A new choice for immunotherapy? European Journal of Medicinal Chemistry 2019, 161, 378-398. [17], Chen, T.; Li, Q.; Liu, Z.; Chen, Y.; Feng, F.; Sun, H. Peptide-based and small synthetic molecule inhibitors on PD-1/PD-L1 pathway: A new choice for immunotherapy? European Journal of Medicinal Chemistry 2019, 161, 378-398.

[18]. Wang, T.; Wu, X.; Guo, C.; Zhang, K.; Xu, J.; Li, Z.; Jiang, S. Development of inhibitors of the programmed cell death-1 /programmed cell death-ligand 1 signaling pathway. Journal of Medicinal Chemistry 2018, 62, 1715-1730. [18]. Wang, T.; Wu, X.; Guo, C.; Zhang, K.; Xu, J.; Li, Z.; Jiang, S. Development of inhibitors of the programmed cell death-1/programmed cell death-ligand 1 signaling pathway. Journal of Medicinal Chemistry 2018, 62, 1715-1730.

[19]. Huck, B.R.; Kótzner, L.; Urbahns, K. Small molecules drive big improvements in immuno-oncology therapies. Angewandte Chemie International Edition 2018, 57, 4412-4428. [19]. Huck, B.R.; Kotzner, L.; Urbahns, K. Small molecules drive big improvements in immuno-oncology therapies. Angewandte Chemie International Edition 2018, 57, 4412-4428.

DESCRIPCIÓN DETALLADA DE LA INVENCIÓN DETAILED DESCRIPTION OF THE INVENTION

Se presenta con fines de invención, un tetradecapéptido cíclico conteniendo un grupo hidracina en la cadena lateral de la lisina para su uso como un nuevo inhibidor de la proteína PD-L1 (¡PD-L1 ), el cual, tras su marcado con "mTc (99mTc-¡PD-L1 ), funciona como un nuevo radiofármaco para la obtención de imágenes de la expresión de PD-L1 . El ligando ¡PD-L1 (-6.7 kcal/mol, afinidad AutoDock) marcado con 99mTc se obtiene con una pureza radioquímica >90%. El radiofármaco [99mTc]Tc-¡PD-L1 puede detectar lesiones tumorales positivas a PD-L1 in vivo en pacientes sometidos a un rastreo de cuerpo entero por tomografía computarizada de emisión de fotón único (SPECT) en medicina nuclear molecular. For the purposes of the invention, a cyclic tetradecapeptide containing a hydrazine group on the lysine side chain is presented for use as a novel inhibitor of the PD-L1 protein (PD-L1), which, after labeling with mTc ( 99mTc -¡PD-L1), functions as a novel radiopharmaceutical for imaging PD-L1 expression. The ¡PD-L1 ligand (-6.7 kcal/mol, AutoDock affinity) labeled with 99mTc is obtained with a radiochemical purity >90%. The radiopharmaceutical [ 99mTc ]Tc-¡PD-L1 can detect PD-L1-positive tumor lesions in vivo in patients undergoing whole-body scanning by single-photon emission computed tomography (SPECT) in molecular nuclear medicine.

Nuestra invención se basa en el uso del grupo hidracina (-NH2-NH2) fuera de un macrociclo peptídico, diferente al WL12 [cyclo(AcTyr-MeAla-Asn-Pro-His-Leu-Hyp-Trp- Ser-Trp(Me)-MeNle-MeNle-Orn-Cys)-Gly-NH2 (puente tioéter -S- entre Cys14 y Ac-Tyr)] al no contener un grupo -S- dentro de la secuencia peptídica del macrociclo, base fundamental de la patente Bristol-Myers Squibb Company, U.S. (Pat. No. 9,879,046). La hidracina como cadena lateral en el macrociclo del tetradecapéptido a patentar denominado hidracina-nicotinoil-PD-L1 [cyclo(Trp-Ser-Trp-Leu-Leu-Lys(hidracina- nicotino¡l)-Cys-Tyr-Ala-Asn-Pro-H¡s-Leu-Pro)] (¡PD-L1 ), genera una doble interacción simultánea, (interacción iónica/puente salino y enlace de hidrógeno) entre el ion carboxilato (-COO-) de la Glu-58 y un ion amonio (-NHs+) de la hidracina presente en el ¡PD-L1 , donde el hidrógeno del -NHs+ forma fuertes enlaces de hidrógeno con el oxígeno de -COO . Es de conocimiento general, que la energía de estabilización debida a las interacciones iónicas (puente salino) es muchas veces superior a la de las interacciones de enlace de hidrógeno, las cuales se suman en una interacción simultánea como la generada en la molécula de ¡PD-L1 (Figura 1). Dicha interacción permitió obtener una estructura más rígida con anclaje y reconocimiento suficiente al ligando PD-L1 sin tener que utilizar, en el nuevo macrociclo peptídico ¡PD-L1 a patentar, sitios de mediación en los aminoácidos, ni enlaces -S- como en la cadena cíclica, como ocurre en el WL12. Además, la Lys proporciona una cadena lateral larga que no interfiriera con el sitio de reconocimiento, mejorando así la afinidad del péptido inhibidor de PD-L1 (¡PD-L1 ) respecto al WL-12 (Tabla 1). Aún tras la conjugación 99mTc-HYNIC (imagen SPECT), la ventaja de la hidracina es su doble grupo amino, por lo que la interacción del amino no terminal con Glu-58 (PD-L1 ) existe dada la cercanía con Glu-58 (4.88 nm)(Figura 1). Método de diseño y preparación del radiooéptido de la invención Our invention is based on the use of the hydrazine group (-NH2-NH2) outside a peptide macrocycle, different from WL12 [cyclo(AcTyr-MeAla-Asn-Pro-His-Leu-Hyp-Trp- Ser-Trp(Me)-MeNle-MeNle-Orn-Cys)-Gly-NH2 (thioether bridge -S- between Cys 14 and Ac-Tyr)] by not containing a -S- group within the peptide sequence of the macrocycle, the fundamental basis of the Bristol-Myers Squibb Company, US patent (Pat. No. 9,879,046). Hydrazine as a side chain in the tetradecapeptide macrocycle to be patented called hydrazine-nicotinoyl-PD-L1 [cyclo(Trp-Ser-Trp-Leu-Leu-Lys(hydrazine-nicotinoyl)-Cys-Tyr-Ala-Asn-Pro-H¡s-Leu-Pro)] (PD-L1 ), generates a simultaneous double interaction (ionic interaction/salt bridge and hydrogen bond) between the carboxylate ion (-COO-) of Glu-58 and an ammonium ion (-NHs + ) of the hydrazine present in the ¡PD-L1 , where the hydrogen of the -NHs + forms strong hydrogen bonds with the oxygen of -COO . It is well known that the stabilization energy due to ionic interactions (salt bridge) is many times higher than that of hydrogen bonding interactions, which add up in a simultaneous interaction such as that generated in the PD-L1 molecule (Figure 1). This interaction made it possible to obtain a more rigid structure with sufficient anchoring and recognition to the PD-L1 ligand without having to use, in the new PD-L1 peptide macrocycle to be patented, mediation sites in the amino acids, or -S- bonds as in the cyclic chain, as occurs in WL12. In addition, Lys provides a long side chain that does not interfere with the recognition site, thus improving the affinity of the PD-L1 inhibitory peptide (PD-L1) with respect to WL-12 (Table 1). Even after 99m Tc-HYNIC conjugation (SPECT image), the advantage of hydrazine is its double amino group, so the interaction of the non-terminal amino with Glu-58 (PD-L1) exists given the proximity to Glu-58 (4.88 nm) (Figure 1). Method of design and preparation of the radiopeptide of the invention

Se utilizó el software AutoDock para realizar el acoplamiento molecular para los cálculos de afinidad. La síntesis química se basó en la reacción de acoplamiento del ácido 6- hidrazinilpiridina-3-carboxílico con un péptido cíclico de 14 aminoácidos. Se preparó ¡PD- L1 para su mareaje con 99mTc. Se utilizó Radio-HPLC para verificar la pureza radioquímica. La estabilidad del radiopéptido en suero humano se evaluó mediante HPLC. La afinidad del [99mTc]Tc-¡PD-L1 se evaluó mediante radio-SDS-PAGE. También se realizó la captación celular de [99mTc]Tc-¡PD-L1 en células PD-L1 positivas (HCC827 y HCT116) y la biodistribución en ratones con tumores inducidos. Un paciente con melanoma maligno plantar avanzado recibió [99mTc]Tc-¡PD-L1 y se realizó un escaneo de cuerpo entero para diferenciar entre las lesiones tumorales (visualizadas por su alta actividad metabólica con [18F]FDG) positivas a PD-L1 (con captación de [99mTc]Tc-¡PD- L1 ) y las lesiones tumorales negativas a PD-L1 (sin captación de [99mTc]Tc-¡PD-L1 ). Acoplamiento molecular (Docking) AutoDock software was used to perform molecular docking for affinity calculations. The chemical synthesis was based on the 6-hydroxybenzoic acid coupling reaction. hydrazinylpyridine-3-carboxylic acid with a 14 amino acid cyclic peptide. ¡PD-L1 was prepared for labeling with 99m Tc. Radio-HPLC was used to verify radiochemical purity. The stability of the radiopeptide in human serum was assessed by HPLC. The affinity of [ 99m Tc]Tc-¡PD-L1 was evaluated by radio-SDS-PAGE. Cellular uptake of [ 99m Tc]Tc-¡PD-L1 in PD-L1 positive cells (HCC827 and HCT116) and biodistribution in tumor-induced mice were also performed. A patient with advanced plantar malignant melanoma received [ 99m Tc]Tc-¡PD-L1 and underwent a whole-body scan to differentiate between PD-L1-positive (with [ 99m Tc]Tc-¡PD- L1 uptake) tumor lesions (visualized by their high metabolic activity with [ 18 F]FDG) and PD-L1-negative (without [ 99m Tc]Tc-¡PD-L1 uptake) tumor lesions. Molecular docking (Docking)

Los ligandos ¡PD-L1 -no conjugado e hidracina-nicotinoil-iPD-L1 , así como WL12 (control positivo) e hidracina-nicotinoil-WL12 (control positivo), se modelaron en dos dimensiones (2D) utilizando ChemDraw para obtener las estructuras básicas. La estructura tridimensional de cada ligando se exportó en formato mol2 utilizando Chem3D para optimizar la geometría molecular utilizando el campo de fuerza MMFF94 a través de la caja de herramientas químicas OpenBabel. Se utilizó como receptor la proteína humana de muerte programada-1 y su ligando (PD-L1 ) obtenida de la base de datos de proteínas (PDB ID: 4ZQK), que se editó para eliminar moléculas de agua, iones y estructuras ajenas a PD-L1 en el proceso de cristalografía de rayos X. Para el cribado virtual, se utilizó cada una de las estructuras mostradas en la Figura 1 como ligando.The unconjugated PD-L1 ligands and hydrazine-nicotinoyl-iPD-L1 , as well as WL12 (positive control) and hydrazine-nicotinoyl-WL12 (positive control), were modeled in two dimensions (2D) using ChemDraw to obtain the basic structures. The three-dimensional structure of each ligand was exported in mol2 format using Chem3D to optimize the molecular geometry using the MMFF94 force field via the OpenBabel chemistry toolbox. Human programmed death-1 protein was used as the receptor and its ligand (PD-L1 ) obtained from the protein database (PDB ID: 4ZQK), which was edited to remove water molecules, ions, and structures unrelated to PD-L1 in the X-ray crystallography process. For virtual screening, each of the structures shown in Figure 1 was used as the ligand.

El receptor como macromolécula y los ligandos correspondientes se prepararon utilizando el paquete gráfico AutoDock Tools 1.5.7, con el cuadro de búsqueda fijado cúbicamente 80 Á en los ejes x, y, y z, centrado en la macromolécula. El acoplamiento semirrígido se realizó con AutoDock vina 1.1.2 alrededor de toda la superficie del receptor, generando 20 poses para cada ligando. Finalmente se generaron las constantes de inhibición (Tabla 1). The receptor as a macromolecule and the corresponding ligands were prepared using the graphical package AutoDock Tools 1.5.7, with the search box cubically fixed 80 Á on the x, y, and z axes, centered on the macromolecule. Semi-rigid docking was performed with AutoDock vina 1.1.2 around the entire receptor surface, generating 20 poses for each ligand. Finally, the inhibition constants were generated (Table 1).

Síntesis y caracterización química Synthesis and chemical characterization

El succinimidil 6-Boc-hidrazinopiridina-3-carboxilato (NHS-HYNIC-Boc) se adquirió a Synchem UG & Co (Felsberg, Alemania). El ciclo(Trp-Ser-Trp-Leu-Leu-Lys-Cys-Tyr-Ala- Asn-Pro-His-Leu-Pro) (PD-L1 -no conjugado) fue ordenado para ser sintetizado a medida de acuerdo con nuestro diseño y de forma exclusiva para nuestro laboratorio por Shanghai Yaxian Chemical Co, Ltd (Jiading, Shanghai, China). Todos los demás reactivos se adquirieron a Merck (Burlington, MA, EE.UU.). Succinimidyl 6-Boc-hydrazinopyridine-3-carboxylate (NHS-HYNIC-Boc) was purchased from Synchem UG & Co (Felsberg, Germany). Cyclo(Trp-Ser-Trp-Leu-Leu-Lys-Cys-Tyr-Ala- Asn-Pro-His-Leu-Pro) (unconjugated PD-L1) was ordered to be custom synthesized according to our design and exclusively for our laboratory by Shanghai Yaxian Chemical Co, Ltd (Jiading, Shanghai, China). All other reagents were purchased from Merck (Burlington, MA, USA).

El péptido hidracina-nicotinoil-iPD-L1 (Figura 2)(6 mg; 3.5 pmol) se disolvió en 0.5 mL de dimetilformamida (DMF), seguido de la adición de N,N-diisopropiletilamina (20 pL). Se disolvió NHS-HYNIC-Boc (2 mg; 5.7 pmol) en 100 pl de DMF y se añadió a la solución peptídica durante 24 h a temperatura ambiente. El NHS-HYNIC-Boc sin reaccionar se eliminó de la mezcla de reacción mediante diálisis (minisistema de diálisis Tube-O- DIALYZER™, 1 kDa MWCO; Merck; Burlington, MA, EE.UU.) durante 24 h utilizando agua inyectable. A la solución acuosa turbia resultante (0,5 mL) se añadió 1 mL de ácido trifluoroacético (TFA) para desproteger el HYNIC (eliminación de Boc), y la mezcla se mantuvo a temperatura ambiente durante 2 h. A continuación, se repitió el proceso de diálisis. Se añadió acetonitrilo a la solución turbia obtenida en una proporción final de 40:60 acetonitrilo: agua para disolver completamente el conjugado. Por último, el conjugado se purificó por HPLC (columna HPLC Discovery® C18 de 5 pm de tamaño de partícula, L x D.l. 25 cm x 10 mm) (Merck; Burlington, MA, EE.UU.) utilizando un gradiente lineal de 0,1 % TFA-agua/0,1% TFA-acetonitrilo del 100 al 20% de la fase acuosa en 20 min a un caudal de 4 mL/min. La fracción recogida entre 13,5 y 15 min se liofilizó. The hydrazine-nicotinoyl-iPD-L1 peptide (Figure 2) (6 mg; 3.5 pmol) was dissolved in 0.5 mL of dimethylformamide (DMF), followed by the addition of N,N-diisopropylethylamine (20 pL). NHS-HYNIC-Boc (2 mg; 5.7 pmol) was dissolved in 100 µl of DMF and added to the peptide solution for 24 h at room temperature. Unreacted NHS-HYNIC-Boc was removed from the reaction mixture by dialysis (Tube-O-DIALYZER™ mini dialysis system, 1 kDa MWCO; Merck; Burlington, MA, USA) for 24 h using injectable water. To the resulting cloudy aqueous solution (0.5 mL), 1 mL of trifluoroacetic acid (TFA) was added to deprotect HYNIC (Boc removal), and the mixture was kept at room temperature for 2 h. The dialysis process was then repeated. Acetonitrile was added to the obtained cloudy solution in a final ratio of 40:60 acetonitrile:water to completely dissolve the conjugate. Finally, the conjugate was purified by HPLC (Discovery® C18 HPLC column, 5 pm particle size, L x Dl 25 cm x 10 mm) (Merck; Burlington, MA, USA) using a linear gradient of 0.1% TFA-water/0.1% TFA-acetonitrile from 100 to 20% of the aqueous phase in 20 min at a flow rate of 4 mL/min. The fraction collected between 13.5 and 15 min was lyophilized.

El polvo resultante se analizó mediante espectroscopia vibracional IR-FT (400-4000 cm-1 , 50 barridos a 0,4 cm-1 ; espectrómetro FT-IR 660, Agilent Technologies). El péptido hidracina-nicotinoil-iPD-L1 disuelto en etanokagua 40:60 se utilizó para su caracterización mediante espectroscopia de masas UPLC (ADQUITY UPLC H-Class con detector de masas QDa; Waters Corporation, Milford, EE.UU.) y espectroscopia UV-vis en el intervalo de 200-400 nm (espectrómetro PerkinElmer LambdaBio; Waltham, Massachusetts, EE.UU.). El espectro de IR y de masas mostraron bandas características vibracionales y masas moleculares correspondientes al péptido hidracina-nicotinoil-iPD- L1 (Figura 3). Radiomarcado The resulting powder was analyzed by FT-IR vibrational spectroscopy (400-4000 cm-1 , 50 scans at 0.4 cm-1 ; FT-IR 660 spectrometer, Agilent Technologies). The peptide hydrazine-nicotinoyl-iPD-L1 dissolved in 40:60 ethane-water was used for characterization by UPLC mass spectroscopy (ADQUITY UPLC H-Class with QDa mass detector; Waters Corporation, Milford, USA) and UV-vis spectroscopy in the range of 200-400 nm (PerkinElmer LambdaBio spectrometer; Waltham, Massachusetts, USA). The IR and mass spectra showed characteristic vibrational bands and molecular masses corresponding to the peptide hydrazine-nicotinoyl-iPD-L1 (Figure 3). Radiolabeled

El péptido hidracina-nicotinoil-iPD-L1 se disolvió en una solución 1 :1 de etanol: agua a una concentración de 1 mg/mL. Para el mareaje con "mTc (formación del complejo [99mTc]Tc-¡PD-L1 ), se disolvieron 100 pL de solución peptídica, 500 pL de ácido et¡lend¡am¡no-N,N'-d¡acét¡co (EDDA)/solución de tricina (30 mg de EDDA en 1 .5 mL de NaOH 0.1 M / 60 mg de tricina en 1 .5 mL de buffer de fosfato 0.2 M a una concentración de 1 mg/mL. 2 M a pH 7) y 500 pL de 99mTcÜ4Na (GETEC, ININ; México) con una actividad de 11 10 MBq, seguido de 20 pL de SnCh (10 mg/10 pL de HCI conc. en 10 mL de agua). La solución se agitó durante 1 min, luego se incubó en un baño seco a 95°C durante 30 min y se dejó enfriar a temperatura ambiente durante 10 min. También se preparó un lote de la formulación liof ilizada que contenía EDDA, tricina, SnCIs y manitol en un área aséptica bajo buenas prácticas de fabricación (área certificada GMP) para su reconstitución con solución de pertecnetato-99m de sodio. Control de calidad The hydrazine-nicotinoyl-iPD-L1 peptide was dissolved in a 1:1 ethanol:water solution at a concentration of 1 mg/mL. For labeling with " m Tc (formation of the complex [ 99m Tc]Tc- ¡PD-L1 ), 100 pL of peptide solution, 500 pL of ethylenediamine-N,N'-diacetic acid (EDDA)/tricine solution (30 mg of EDDA in 1 .5 mL of 0.1 M NaOH / 60 mg of tricine in 1 .5 mL of 0.2 M phosphate buffer at a concentration of 1 mg / mL. 2 M at pH 7) and 500 pL of 99m TcÜ4Na (GETEC, ININ; Mexico) with an activity of 11 10 MBq were dissolved, followed by 20 pL of SnCh (10 mg/10 pL of conc. HCl in 10 mL of water). The solution was stirred for 1 min, then incubated in a dry bath at 95°C for 30 min and allowed to cool to room temperature over 10 min. A batch of the lyophilized formulation containing EDDA, tricine, SnCls and mannitol was also prepared in an aseptic area under good manufacturing practices (GMP certified area) for reconstitution with sodium pertechnetate-99m solution. Quality control

Para el control de calidad de la formulación [99mTc]Tc-¡PD-L1 , se analizaron parámetros como el aspecto (solución clara), pH (neutro), esterilidad, ausencia de endotoxinas bacterianas y pureza radioquímica (radio-HPLC; Waters Corporation, Milford, USA; 3. 9 mm x 30 cm pBondapak™ , columna C18; sistema lineal de gradientes; solvente A: 0.1 % TFA-acetonitrilo y solvente B: 0.1 % TFA-agua, de 100 to 30% de A en 20 min) se evaluaron de acuerdo con la Farmacopea Mexicana [16], en su sección de "Métodos Generales de Análisis" (MGA). El tiempo de retención de [99mTc]Tc-¡PD-L1 fue de 16.5±0.3 min (Figura 3), mientras que los tiempos de retención de 99mTcO4_ y [99mTc]Tc-EDDA/tric¡na fueron de 3.5 ± 0.2 min y 4.5 ± 0.2 min, respectivamente, confirmando una pureza radioquímica mayor al 95% para el [99mTc]Tc-¡PD-L1 (Figura 3). Estabilidad For quality control of the [ 99m Tc]Tc-¡PD-L1 formulation, parameters such as appearance (clear solution), pH (neutral), sterility, absence of bacterial endotoxins, and radiochemical purity (radio-HPLC; Waters Corporation, Milford, USA; 3.9 mm x 30 cm pBondapak™, C18 column; linear gradient system; solvent A: 0.1% TFA-acetonitrile and solvent B: 0.1% TFA-water, from 100 to 30% of A in 20 min) were evaluated according to the Mexican Pharmacopoeia [16], in its "General Methods of Analysis" (MGA) section. The retention time of [ 99m Tc]Tc-¡PD-L1 was 16.5 ± 0.3 min (Figure 3), while the retention times of 99m TcO4 _ and [ 99m Tc]Tc-EDDA/tricin were 3.5 ± 0.2 min and 4.5 ± 0.2 min, respectively, confirming a radiochemical purity greater than 95% for [ 99m Tc]Tc-¡PD-L1 (Figure 3). Stability

La estabilidad de [99mTc]Tc-¡PD-L1 se confirmó diluyendo muestras del radiocomplejo en suero humano (5x). Los conjugados (n=3) se incubaron a 37°C, luego se tomaron muestras a las 0.5, 3 y 24 h, y se evaluó la pureza radioquímica mediante radio-HPLC de exclusión molecular (ProteinPak 300SW, Waters Corporation, Milford, USA). Los estudios de estabilidad in vitro de [99mTc]Tc-ÍPD-L1 en suero humano mostraron una unión a proteínas séricas del 1 .3±0.8% a los 30 min, de 3.2±1 .3% a las 3 h y del 7.6±1 .9% a las 24 h, así como una alta estabilidad radioquímica (> 90% a las 24 h). Reconocimiento molecular por la proteína PD-L1 : radio-SDS-PAGE The stability of [ 99m Tc]Tc-PD-L1 was confirmed by diluting radiocomplex samples in human serum (5x). Conjugates (n = 3) were incubated at 37°C, then samples were taken at 0.5, 3 and 24 h, and radiochemical purity was assessed by size exclusion radio-HPLC (ProteinPak 300SW, Waters Corporation, Milford, USA). In vitro stability studies of [ 99m Tc]Tc-PD-L1 in human serum showed serum protein binding of 1.3 ± 0.8% at 30 min, 3.2 ± 1.3% at 3 h and 7.6 ± 1.9% at 24 h, as well as high radiochemical stability (> 90% at 24 h). Molecular recognition by the PD-L1 protein: radio-SDS-PAGE

Se realizó una electroforesis en gel de dodecil sulfato sódico-poliacrilamida (SDS- PAGE). La distribución de la radiactividad se detectó utilizando un escáner de radio-TLC (Mini-Gita, 60-150 keV, BGO-Crystal, ventana 25 x 2 mm, espesor 5 mm, resolución optimizada para Tc-99m; RayTest, Munich, Alemania) para detectar la unión de [99mTc]Tc-ÍPD-L1 a la proteína PD-L1 . Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was performed. The distribution of radioactivity was detected using a radio-TLC scanner (Mini-Gita, 60-150 keV, BGO-Crystal, window 25 x 2 mm, thickness 5 mm, resolution optimized for Tc-99m; RayTest, Munich, Germany) to detect the binding of [ 99m Tc]Tc-IPD-L1 to the PD-L1 protein.

Brevemente, se incubó una solución de ¡PD-L1 marcadas con 99mTc (100pL) (1 mg/ml de péptido) con proteína PD-L1 recombinante humana (100pL) (200 pg/mL; FineTest; Cat: P6857;)(interacción radiopéptido-PD-L1 ) o proteína humana integrina aVp3 (100pL) (control negativo; 200 pg/mL) (Chemicom, Merck; Burlington, MA, USA) (interacción radiopéptido-integrina) durante 30 min a 37°C, tras lo cual las muestras se diluyeron con tampón de muestra Laemmli (conteniendo 10% SDS, Tris-CI 125 mM, pH = 6. 8, 12.5% de glicerol y 0.005% de azul de bromofenol después del colorante) para dar concentraciones finales de proteína de 25 pg/mL. Las muestras (99mTc-¡PD-L1 , proteína PD-L1 , integrina, proteína 99mTc-¡PD-L1 -PD-L1 y 99mTc-¡PD-L1 -integrina) se cargaron en el gel SDS-PAGE a temperatura ambiente. Se cargaron 30 pL (30 pg) de cada muestra en pocilios de 1 .5 mm de espesor de geles de poliacrilamida al 15%. Los experimentos se realizaron en un solo gel. En todos los casos, la electroforesis se ejecutó hasta justo antes de que el colorante de bromofenol alcanzara el fondo del gel (distancia total del gel de 100 mm). En cada gel se utilizaron también patrones de peso molecular preestablecidos (BLUE stainTM Protein Ladder 11 -245 kDa Cat: P007-500). Los geles se tiñeron con azul Coomassie de 30 a 45 minutos a temperatura ambiente y se dejaron decolorar de 4 a 20 horas antes de fotografiarlos. Finalmente, la distribución de la radiactividad en los geles se escaneó utilizando un detector de radio-TLC para determinar el porcentaje de radiactividad [99mTc]Tc-¡PD-L1 asociada a las proteínas. Los resultados mostraron 30% de la radiactividad [99mTc]Tc-¡PD-L1 asociada al reconocimiento molecular de PD-L1 (Figura 4). Briefly, a solution of 99m Tc-labeled PD-L1 (100 µL) (1 mg/mL peptide) was incubated with recombinant human PD-L1 protein (100 µL) (200 pg/mL; FineTest; Cat: P6857;)(radiopeptide-PD-L1 interaction) or human integrin αVp3 protein (100 µL) (negative control; 200 pg/mL) (Chemicom, Merck; Burlington, MA, USA) (radiopeptide-integrin interaction) for 30 min at 37°C, after which the samples were diluted with Laemmli sample buffer (containing 10% SDS, 125 mM Tris-Cl, pH = 6.8, 12.5% glycerol and 0.005% bromophenol blue after dye) to give final protein concentrations. of 25 pg/mL. Samples ( 99m Tc-¡PD-L1, PD-L1 protein, integrin, 99m Tc-¡PD-L1 protein -PD-L1 and 99m Tc-¡PD-L1 -integrin) were loaded onto SDS-PAGE gel at room temperature. 30 pL (30 pg) of each sample were loaded into 1.5 mm thick wells of 15% polyacrylamide gels. The experiments were performed on a single gel. In all cases, electrophoresis was run until just before the bromophenol dye reached the bottom of the gel (total gel distance of 100 mm). Pre-established molecular weight standards (BLUE stainTM Protein Ladder 11-245 kDa Cat: P007-500) were also used on each gel. The gels were stained with Coomassie blue for 30 to 45 minutes at room temperature and allowed to destain for 4 to 20 hours before being photographed. Finally, the distribution of radioactivity in the gels was scanned using a radio-TLC detector to determine the percentage of [ 99m Tc]Tc-¡PD-L1 radioactivity associated with proteins. The results showed 30% of the [ 99m Tc]Tc-¡PD-L1 radioactivity associated with molecular recognition of PD-L1 (Figure 4).

Inmunofluorescencia Immunofluorescence

La expresión celular de PD-L1 se confirmó mediante inmunofluorescencia. Las líneas celulares HCC827, HCT116 y C6 se fijaron con paraformaldehído al 4% durante 20 minutos, se permeabilizaron con TritonX-100 al 0,5% y se bloquearon con albúmina de suero bovino al 1%. A continuación, las células se incubaron durante la noche con un anticuerpo anti-PD-L1 según las instrucciones del proveedor (FineTest, Wuhan Fine Biotech Co., China). A continuación, se incubaron las células con IgG anti-conejo de cabra conjugado con Alexa Fluor 488 (H+L) (Invitrogen Cat. No. A32731 ) durante 1 h. Se utilizó DAPI para observar la intensidad de fluorescencia en las células para la tinción nuclear. La fluorescencia se observó mediante microscopía (Meiji Techno; modelo MT6200; Saitama, Japón). Cellular PD-L1 expression was confirmed by immunofluorescence. HCC827, HCT116, and C6 cell lines were fixed with 4% paraformaldehyde for 20 min, permeabilized with 0.5% TritonX-100, and blocked with 1% bovine serum albumin. Cells were then incubated overnight with an anti-PD-L1 antibody according to the supplier's instructions (FineTest, Wuhan Fine Biotech Co., China). Cells were then incubated with Alexa Fluor 488-conjugated goat anti-rabbit IgG (H+L) (Invitrogen Cat. No. A32731) for 1 h. DAPI was used to observe the fluorescence intensity in cells for nuclear staining. Fluorescence was observed by microscopy (Meiji Techno; model MT6200; Saitama, Japan).

Captación e internalización celular Cellular uptake and internalization

Las células diluidas en solución salina tamponada con fosfato (PBS) (pH 7,4) (1 x106 células/tubo) recibieron dos tratamientos diferentes: a) [99mTc]Tc-¡PD-L1 (7.4 kBq) (n=3), y b) 99mTcO4Na (7.4 kBq) (n=3). Las células se incubaron con cada tratamiento a 37 °C durante 1 h. Tras la incubación, los tubos se midieron en un detector de Nal(TI) (NML Inc., Houston, Texas, EE.UU.) para determinar la actividad inicial (100%). Los tubos se centrifugaron a 500 g durante 10 minutos, se lavaron una vez con PBS y se volvieron a centrifugar. Se eliminó el líquido y se midió la actividad de botón, correspondiente al porcentaje de captación celular (captación superficial) con respecto a la actividad inicial. Se añadió una mezcla de ácido acético/0.5 M NaCI, y se repitió la centrifugación, seguida de la medición de la actividad. Se eliminó el líquido y se midió la actividad de botón, correspondiente al porcentaje de actividad internalizada por las células con respecto a la actividad de captación (fracción de internalización de la captación superficial). El porcentaje de captación celular del [99mTc]Tc-¡PD-L1 correlacionó con la expresión de PD-L1 celular determinado por inmunoflurescencia en cada una de las líneas celulares analizadas. La captación fue nula para 99mTcO4Na (control negativo) en todas las células analizadas. Cells diluted in phosphate-buffered saline (PBS) (pH 7.4) (1 x 106 cells/tube) received two different treatments: a) [ 99m Tc]Tc-¡PD-L1 (7.4 kBq) (n = 3), and b) 99m TcO4Na (7.4 kBq) (n = 3). Cells were incubated with each treatment at 37 °C for 1 h. After incubation, tubes were measured in a Nal(TI) detector (NML Inc., Houston, Texas, USA) to determine the initial activity (100%). The tubes were centrifuged at 500 g for 10 minutes, washed once with PBS, and centrifuged again. The liquid was removed, and the button activity, corresponding to the percentage of cellular uptake (surface uptake) with respect to the initial activity, was measured. A mixture of acetic acid/0.5 M NaCl was added, and centrifugation was repeated, followed by activity measurement. The liquid was removed, and the button activity, corresponding to the percentage of activity internalized by the cells with respect to the uptake activity (fraction of internalization of surface uptake), was measured. The percentage of cellular uptake of [ 99m Tc]Tc-¡PD-L1 correlated with the expression of Cellular PD-L1 was determined by immunofluorescence in each of the cell lines analyzed. Uptake was zero for 99m TcO4Na (negative control) in all cells analyzed.

Biodistribución Biodistribution

Todos los procedimientos con animales se realizaron de acuerdo con el Reglamento Ético para el Manejo de Animales de Laboratorio (NOM-062-ZOO-1999) y los requerimientos del Comité Institucional para el Cuidado y Uso de Animales bajo un protocolo aprobado (No. 09-2018-2022). Los ratones machos Nu/Nu (CINVESTAV, I.PN., Ciudad de México), de 6-8 semanas de edad, se mantuvieron en un ambiente de barrera aséptica. Los ratones fueron inoculados mediante inyección subcutánea de 1 x106 células HCC827/0,1 mL PBS en la parte superior de la espalda. Los lugares de inyección se controlaron periódicamente para detectar la progresión tumoral. El día 10 después de la inoculación de las células tumorales, se inyectó a los animales 18.5 MBq (50 pL) de [99mTc]Tc-ÍPD-L1 en la vena de la cola. Los animales fueron sacrificados a las 1 , 3 y 24 h (n=3). Se diseccionaron muestras de riñones, pulmones, corazón, hígado, bazo, páncreas, intestino delgado, estómago, músculo, sangre y tumor. La radiactividad se midió en un detector de Nal(TI). Los resultados se expresaron como porcentaje de la dosis inyectada por gramo de tejido (%ID/g), mostrando captación de [99mTc]Tc-ÍPD-L1 en los tumores positivos a ¡PD-L1 (Figura 5). All animal procedures were performed in accordance with the Ethical Regulations for the Management of Laboratory Animals (NOM-062-ZOO-1999) and the requirements of the Institutional Committee for the Care and Use of Animals under an approved protocol (No. 09-2018-2022). Male Nu/Nu mice (CINVESTAV, I.PN., Mexico City), 6-8 weeks old, were maintained in an aseptic barrier environment. Mice were inoculated by subcutaneous injection of 1 x106 HCC827 cells/0.1 mL PBS into the upper back. Injection sites were periodically monitored for tumor progression. On day 10 after tumor cell inoculation, animals were injected into the tail vein with 18.5 MBq (50 pL) of [ 99m Tc]Tc-ÍPD-L1. Animals were sacrificed at 1, 3, and 24 h (n=3). Samples of kidneys, lungs, heart, liver, spleen, pancreas, small intestine, stomach, muscle, blood, and tumor were dissected. Radioactivity was measured using a Nal(TI) detector. Results were expressed as percentage of the injected dose per gram of tissue (%ID/g), showing uptake of [ 99m Tc]Tc-IPD-L1 in the ipD-positive tumors (Figure 5).

Imagen clínica Clinical image

Se muestra un estudio de un hombre de 62 años (peso 60 kg; altura 168 cm) diagnosticado de melanoma maligno plantar avanzado (Comité de Ética: Protocolo ne 2023-MN02). El estudio se aprobó basándose en el concepto de microdosis y en estudios de prueba de concepto. Además, se tuvieron en cuenta las normas éticas del Hospital INCan basadas en la Declaración de Helsinki relativas a la experimentación en seres humanos y el certificado GMP concedido al IN IN por la Secretaría de Salud de México. El paciente firmó un consentimiento informado tras recibir información detallada sobre el propósito del estudio, que podría ayudar a decidir el tratamiento y a monitoñzar la progresión de la enfermedad. El paciente fue sometido a SPECT/CT (Symbia TruePoint, Siemens) 2 horas después de la administración intravenosa de [99mTc]Tc-ÍPD-L1 (740 MBq) y había tenido una exploración previa de [18F]FDG PET/CT (Excel 20; Siemens Medical Solutions) 5 días antes. Se realizó un escaneo de cuerpo entero para diferenciar entre las lesiones tumorales (todas aquellas visualizadas por su alta actividad metabólica con [18F]FDG) positivas a PD-L1 (captación de [99mTc]Tc-¡PD-L1 ) y las lesiones tumorales negativas a PD-L1 (sin captación de [99mTc]Tc-ÍPD-L1 ) (Figura 6). Concluyendo, el [99mTc]Tc-iPD-L1 se obtiene con las siguientes características: A study of a 62-year-old man (weight 60 kg; height 168 cm) diagnosed with advanced plantar malignant melanoma is shown (Ethics Committee: Protocol n e 2023-MN02). The study was approved based on the microdose concept and proof-of-concept studies. In addition, the ethical standards of INCan Hospital based on the Declaration of Helsinki regarding human experimentation and the GMP certificate granted to INCan Hospital by the Mexican Ministry of Health were taken into account. The patient signed an informed consent after receiving detailed information about the purpose of the study, which could help decide on the treatment and monitor disease progression. The patient underwent SPECT/CT (Symbia TruePoint, Siemens) 2 hours after intravenous administration of [ 99m Tc]Tc-IPD-L1 (740 MBq) and had had a previous [ 18 F]FDG PET/CT scan (Excel 20; Siemens Medical Solutions) 5 days earlier. A whole-body scan was performed to differentiate between PD-L1-positive ([ 99m Tc]Tc-IPD-L1 uptake) tumor lesions (all those visualized by their high metabolic activity with [ 18 F]FDG) and PD-L1-negative (no [ 99m Tc]Tc-IPD-L1 uptake) tumor lesions (Figure 6). In conclusion, the [ 99m Tc]Tc-iPD-L1 is obtained with the following characteristics:

• Pureza radioquímica mayor al 95 % • Radiochemical purity greater than 95%

• Capacidad del radiofármaco para detectar in vivo y de forma específica, a tumores que sobre-expresan a la proteína de punto de control inmunitario PD-L1 por tomografía de emisión de fotón único (SPECT) en medicina nuclear. • Ability of the radiopharmaceutical to detect in vivo and specifically tumors that overexpress the immune checkpoint protein PD-L1 by single photon emission computed tomography (SPECT) in nuclear medicine.

• Además del reconocimiento molecular de la secuencia de aminoácidos de tetradecapéptido marcado con 99mTc, el [99mTc]Tc-¡PD-L1 tiene la habilidad de captarse significativamente y detectar con alta sensibilidad a tumores y metástasis con expresión PD-L1 , debido una doble interacción simultánea, (interacción iónica/puente salino y enlace de hidrógeno) entre el ion carboxilato (-COO-) de la Glu-58 y un ion amonio (- NHs+) de la hidracina presente en el ¡PD-L1 , donde el hidrógeno del -NHs+ forma fuertes enlaces de hidrógeno con el oxígeno de -COO . Es de conocimiento general, que la energía de estabilización debida a las interacciones iónicas (puente salino) es muchas veces superior a la de las interacciones de enlace de hidrógeno, las cuales se suman en una interacción simultánea como la generada en la molécula de ¡PD-L1 . • In addition to the molecular recognition of the 99mTc -labeled tetradecapeptide amino acid sequence, [ 99mTc ]Tc-¡PD-L1 has the ability to significantly uptake and detect with high sensitivity tumors and metastases with PD-L1 expression, due to a simultaneous double interaction (ionic/salt bridge interaction and hydrogen bonding) between the carboxylate ion (-COO-) of Glu-58 and an ammonium ion (-NHs + ) of the hydrazine present in ¡PD-L1 , where the hydrogen of -NHs + forms strong hydrogen bonds with the oxygen of -COO . It is general knowledge that the stabilization energy due to ionic interactions (salt bridge) is many times higher than that of hydrogen bonding interactions, which add up to a simultaneous interaction such as that generated in the ¡PD-L1 molecule.

Claims

REIVINDICACIONES Habiendo descrito suficientemente mi invención, considero como una novedad y por lo tanto reclamo como de mi exclusiva propiedad, lo contenido en las siguientes claúsulas: Having sufficiently described my invention, I consider as a novelty and therefore claim as my exclusive property, the contents of the following clauses: 1 Un compuesto que tiene la estructura hidracina-nicotinoil-PD-L1 [cyclo(Trp-Ser- Trp-Leu-Leu-Lys(hidracina-nicotinoil)-Cys-Tyr-Ala-Asn-Pro-His-Leu-Pro)] (¡PD-L1 ):
Figure imgf000015_0001
1 A compound having the structure hydrazine-nicotinoyl-PD-L1 [cyclo(Trp-Ser-Trp-Leu-Leu-Lys(hydrazine-nicotinoyl)-Cys-Tyr-Ala-Asn-Pro-His-Leu-Pro)] (PD-L1):
Figure imgf000015_0001
2.- Un compuesto que tiene la estructura que se reclama en la reivindicación 1 en el que se sustituye la Lys(hidracina-nicotinoil) por cualquier molécula dedicada a quelar un radionúclido. 2.- A compound having the structure claimed in claim 1 in which Lys(hydrazine-nicotinoyl) is replaced by any molecule dedicated to chelating a radionuclide. 3.- Un radiofármaco con la fórmula: [99mTc]Tc-¡PD-L1 teniendo la siguiente estructura: 3.- A radiopharmaceutical with the formula: [ 99m Tc]Tc-PD-L1 having the following structure:
Figure imgf000016_0001
Figure imgf000016_0001
4.- Un radiofármaco como el que se reclama en la reivindicación 3 para ser usado4.- A radiopharmaceutical as claimed in claim 3 to be used 410 en la detección de tumores que expresan la proteína PD-L1 . 410 in the detection of tumors expressing the PD-L1 protein.
PCT/MX2024/050024 2023-11-16 2024-04-08 [99mtc]tc-ipd-l1 as a tetradecapeptide radiopharmaceutical for detecting and monitoring the overexpression of the pd-l1 protein Pending WO2025105948A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
MXMX/A/2023/013591 2023-11-16
MX2023013591 2023-11-16

Publications (1)

Publication Number Publication Date
WO2025105948A1 true WO2025105948A1 (en) 2025-05-22

Family

ID=95743184

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/MX2024/050024 Pending WO2025105948A1 (en) 2023-11-16 2024-04-08 [99mtc]tc-ipd-l1 as a tetradecapeptide radiopharmaceutical for detecting and monitoring the overexpression of the pd-l1 protein

Country Status (1)

Country Link
WO (1) WO2025105948A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018085750A2 (en) * 2016-11-07 2018-05-11 Bristol-Myers Squibb Company Immunomodulators
CN116983441A (en) * 2023-06-07 2023-11-03 复旦大学附属肿瘤医院 A kind of cyclic peptide radiopharmaceutical for targeting PD-L1 and its preparation method and use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018085750A2 (en) * 2016-11-07 2018-05-11 Bristol-Myers Squibb Company Immunomodulators
CN116983441A (en) * 2023-06-07 2023-11-03 复旦大学附属肿瘤医院 A kind of cyclic peptide radiopharmaceutical for targeting PD-L1 and its preparation method and use

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
FERRO-FLORES GUILLERMINA, OCAMPO-GARCÍA BLANCA, CRUZ-NOVA PEDRO, LUNA-GUTIÉRREZ MYRNA, BRAVO-VILLEGAS GERARDO, AZORÍN-VEGA ERIKA, : "99mTc-Labeled Cyclic Peptide Targeting PD-L1 as a Novel Nuclear Imaging Probe", PHARMACEUTICS, MDPI AG, SWITZERLAND, vol. 15, no. 12, Switzerland, pages 2662, XP093232339, ISSN: 1999-4923, DOI: 10.3390/pharmaceutics15122662 *
GE, S. ET AL.: "A novel 68Ga-labeled cyclic peptide molecular probe based on the computer-aided design for noninvasive imaging of PD-L1 expression in tumor s", BIOORGANIC CHEMISTRY, vol. 140, 23 August 2023 (2023-08-23), XP087399194, ISSN: 0045-2068, Retrieved from the Internet <URL:https://pubmed.ncbi.nlm.nih.gov/37639759> [retrieved on 20240718], DOI: 10.1016/j.bioorg.2023.106785 *
LIU, S. ET AL.: "A Novel Ternary Ligand System for 99mTc-Labeling of Hydrazino Nicotinamide-Modified Biologically Active Molecules Using Imine-N-Containing Heterocycles as Coligands", BIOCONJUGATE CHEMISTRY, vol. 9, no. 5, - 15 August 1998 (1998-08-15), pages 583 - 595, XP000778001, ISSN: 1043-1802, Retrieved from the Internet <URL:https://pubmed.ncbi.nlm.nih.gov/9736492> [retrieved on 20240719], DOI: 10.1021/bc9800116 *
LUNA-GUTIÉRREZ MYRNA, CRUZ-NOVA PEDRO, JIMÉNEZ-MANCILLA NALLELY, OROS-PANTOJA RIGOBERTO, LARA-ALMAZÁN NANCY, SANTOS-CUEVAS CLARA, : "Synthesis and Evaluation of 177Lu-DOTA-PD-L1-i and 225Ac-HEHA-PD-L1-i as Potential Radiopharmaceuticals for Tumor Microenvironment-Targeted Radiotherapy", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, MOLECULAR DIVERSITY PRESERVATION INTERNATIONAL (MDPI), BASEL, CH, vol. 24, no. 15, Basel, CH , pages 12382, XP093135761, ISSN: 1422-0067, DOI: 10.3390/ijms241512382 *
MISHRA, A. ET AL.: "Gallium-68-labeled Peptide PET Quantifies Tumor Exposure of PD-L1 Therapeutics", CLINICAL CANCER RESEARCH, vol. 29, no. 3, - 30 November 2022 (2022-11-30), pages 581 - 591, XP093232247, ISSN: 1078-0432, Retrieved from the Internet <URL:https://aacrjournals.org/clincancerres/article/29/3/581/716099/Gallium-68-labeled-Peptide-PET-Quantifies-Tumor> [retrieved on 20240719], DOI: 10.1158/1078-0432.CCR-22-1931 *

Similar Documents

Publication Publication Date Title
ES2844586T3 (en) 18F-tagged prostate specific membrane antigen (PSMA) inhibitors and their use as imaging agents for prostate cancer
ES2864091T3 (en) Positron Emission Tomography (PET) Imaging Immunomodulators
ES2972124T3 (en) Peptides labeled with a positron-emitting radionuclide for PET imaging of human uPAR
US10464985B2 (en) Compounds with reduced ring size for use in diagnosing and treating melanoma, including metastatic melanoma and methods related to same
ES2960729T3 (en) 177 Lu-dota-hynic-ipsma as a therapeutic radiopharmaceutical targeting prostate-specific membrane antigen
US12465660B2 (en) Compositions and methods for cancer imaging and radiotherapy
CN111630059A (en) Novel Radiometal Binding Compounds for Diagnosis or Treatment of Cancers Expressing Prostate Specific Membrane Antigen
ES2794581T3 (en) Procedures and kits for preparing radionuclide complexes
KR20120050462A (en) Folate-targeted diagnostics and treatment
US11141494B2 (en) Development of neurokinin-1 receptor-binding agent delivery conjugates
Li et al. 188Re-labeled hyperbranched polysulfonamine as a robust tool for targeted cancer diagnosis and radioimmunotherapy
Lindeman et al. FAP Radioligand Linker Optimization improves Tumor Dose and Tumor-to-healthy organ ratios in 4T1 syngeneic model
WO2025105948A1 (en) [99mtc]tc-ipd-l1 as a tetradecapeptide radiopharmaceutical for detecting and monitoring the overexpression of the pd-l1 protein
CN117813326A (en) Radiation-Based Detection, Companion Testing, and Treatment of Directin-1
KR101551232B1 (en) Novel N3S1 chelator-folate derivatives, preparation method thereof and composition for diagnosis or treatment of cancer containing the same as an active ingredient
WO2025171887A1 (en) Radiolabeled complexes and pharmaceutical compositions including the same
Ahmad The Use of Bioorthogonal Chemistry to Create 99mTc and Micelle-based Diagnostic Agents
Ngo Integrating Metabolic, Neo-Angiogenic, and HER2-Targeted Imaging to Characterize Breast Cancer in Preclinical Models
WO2024206972A1 (en) Compounds and combinations for diagnosing and treating melanoma and methods related to the same
WO2025191096A1 (en) Bicyclic peptide
Li Metal chelating agents for medicinal radionuclides
EP4612189A1 (en) Radionuclide and near-infrared dye conjugated antibody for detection of gd2-positive cancer
Bilton THE SYNTHESIS AND EVALUATION OF NEW RADIOPHARMACEUTICALS AND MULTIMODAL IMAGING PROBES
US20100104511A1 (en) Methods and compositions using chelator-antibody conjugates
BR112017010414B1 (en) 18F-RADIOLABELED PROTEIN-BASED PROBES AND PHARMACEUTICAL COMPOSITIONS

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24891892

Country of ref document: EP

Kind code of ref document: A1