CN110227169A - A kind of nuclear medicine drug of the rgd peptide of structural modification - Google Patents

A kind of nuclear medicine drug of the rgd peptide of structural modification Download PDF

Info

Publication number
CN110227169A
CN110227169A CN201910441556.7A CN201910441556A CN110227169A CN 110227169 A CN110227169 A CN 110227169A CN 201910441556 A CN201910441556 A CN 201910441556A CN 110227169 A CN110227169 A CN 110227169A
Authority
CN
China
Prior art keywords
dota
3prgd
rgd
bfc
carboxyl
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.)
Granted
Application number
CN201910441556.7A
Other languages
Chinese (zh)
Other versions
CN110227169B (en
Inventor
王凡
史继云
贾兵
高瀚男
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.)
Peking University
Original Assignee
Peking University
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 Peking University filed Critical Peking University
Priority to CN201910441556.7A priority Critical patent/CN110227169B/en
Publication of CN110227169A publication Critical patent/CN110227169A/en
Priority to PCT/CN2020/091861 priority patent/WO2020238800A1/en
Priority to EP20815331.2A priority patent/EP3906947A4/en
Priority to US17/310,604 priority patent/US20230117927A1/en
Application granted granted Critical
Publication of CN110227169B publication Critical patent/CN110227169B/en
Active 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
    • A61K51/082Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins the peptide being a RGD-containing peptide
    • 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

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A kind of rgd peptide of structural modification, the complex formed by the polypeptide and radionuclide, and the pharmaceutical composition comprising the complex, described pharmaceutical composition is for diagnosing or treating integrin alpha v beta 3 positive tumor.The complex, which has, such as gives a definition: wherein A is such as flowering structure to A- (L) n-RGD polypeptide:L represents linking arm molecule, has the following structure:Wherein m is the integer of 1-8.

Description

A kind of nuclear medicine drug of the rgd peptide of structural modification
Technical field
The nuclear medicine drug more particularly to structural modification RGD modified the present invention relates to a kind of pair of cancer target rgd peptide are more The diagnosis or treatment radiopharmaceutical of peptide.
Background technique
Molecular image refers to that image application carries out the qualitative of cell and molecular level to the bioprocess under condition of living organism And quantitative study.Molecular probe be the key that molecular imaging development, to a certain specific biological molecules (such as protein, RNA, DNA) there are labeled compound molecule that is selectively targeted and being able to carry out internal or external tracer, these labeled compounds Molecule is able to reflect the amount function of its targeting biological molecules.Molecular probe can be divided into core according to the difference of imaging means used Medical probe, optical probe and MRI probe etc., wherein nuclear medicine molecular probe has unique advantage.Molecular probe according to Molecular size range classification includes small molecule class, and the size of polypeptide, antibody class, the types such as nano particle, molecular weight can very great Cheng The kinetic property of drug is influenced on degree, wherein the tumor targeted molecular probe of small-molecular-weight has shown many unique excellent Point, is widely studied and applied, such as the RGD class polypeptide of targeted integration element α v β 3.
The continued propagation of malignant tumour, invasion, transfer depend on Tumor Angiongesis.Integrin is to participate in modulate tumor blood One of the important factor of pipe hyperplasia, plays an important role to Tumor Angiongesis, in integrin most with the effect of integrin alpha v beta 3 It is important.Integrin alpha v beta 3 is expressed in endothelial cells in tumor neogenetic blood vessels height, and is not expressed in normal cell or mature blood vessel Or low expression.RGD can specifically bind integrin alpha v beta 3, therefore be set using the specific binding of RGD and integrin alpha v beta 3 The RGD class molecular probe counted out obtains widely studying and using.Domestic and international researcher, which successively passes through, is transformed cyclization for linear RGD Shape RGD, pharmacokinetics bridging agent modification for example RGD is glycosylated, with glutamic acid by RGD cyclic peptide monomer be connected into polymer, Optimize RGD by being inserted into Gly3 or PEG4 chain etc. a variety of methods between the two RGD monomers of chemical means in dimer Polypeptide, to obtain the rgd peptide of structural modification.
However, many ring-type RGD monomeric peptide radioactively labelled substances, tumor uptake is low, and serum removal rates are fast, the devices such as kidney, liver Official absorbs height, these all limit application of the cyclic monomer peptide as imaging agent;As multimerization degree is promoted, radioactivity RGD Polypeptide also obviously increases in the intake of the organs such as kidney, liver, lung, moreover, the higher synthesis of multimerization degree is more complicated, cost is higher, this A little is also the restraining factors of RGD probe multimerization development;Simple multimerization or the pharmacokinetics bridging agent using macromolecule The advantage modified RGD probe is no longer obvious.
Summary of the invention
In order to improve the above problem of the existing technology, the present invention provides the rgd peptide such as following formula structural modification:
A- (L) n-RGD polypeptide
Wherein A is such as flowering structure:
L represents linking arm molecule, has the following structure:Wherein m is the integer of 1-8, such as 2-6, preferably 5;
The L is by the amino reaction key chain in its carboxyl and A, for example, by marking the carboxyl and A key chain that are in L;
N is 0 or 1;
When n is 0, the rgd peptide passes through the carboxyl reaction key chain in its amino and A;
When n is 1, the rgd peptide is by another carboxyl reaction key chain in its amino and L, such as when marking in L When being connected for the carboxyl of * with A, labeled as the carboxyl and rgd peptide reaction key chain of * *;
Rgd peptide is rgd peptide selected from the following: c (RGDfV), c (RGDfK), c (RGDfE), c (RGDyk), E [c (RGDyk)]2、E[c(RGDfK)]2、3PRGD2
The present invention also provides the complexs of the radioisotope labeling of the rgd peptide containing structural modification as above, have Such as undefined structure:
Nu-BFC-A- (L) n-RGD polypeptide
Wherein:
Nu is radionuclide, such as diagnostic imaging nucleic:111In、64Cu、99mTc、68Ga, or treatment nucleic:90Y、177Lu、89Sr、153Sm、188Re;BFC is bifunctional chelating agent (bifunctional chelating agent), such as HYNIC (buzane niacinamide), MAG2(mercaptoacetyl Diglycocol), MAG3(mercaptoacetyl triglycine), DTPA (diethyl triamine five Acetic acid), DOTA (- 1,4,7,10 tetraacethyl of Cyclen), NOTA (7-triazacyclononane -1 1,4,7-, 4,7 tricarboxylic acids), TETA (- 1,4,8,11 tetraacethyl of 1,4,8,11- tetraazacyclododecane tetradecane);
When n is 0, the bifunctional chelating agent passes through the-NH in the carboxyl and A having in its structure2Reaction key chain;
When n is 1, the bifunctional chelating agent passes through the-NH in the carboxyl and L having in its structure2Reaction key chain.
According to the present invention, in the polypeptide RGD and complex of structural modification as above:
Rgd peptide is selected from: c (RGDfK), 3PRGD2
Nu is selected from:90Y、177Lu、111In、64Cu、99mTc;
When Nu is90Y、177When Lu, BFC is selected from DTPA, DOTA;When Nu is111In、64Cu、68Ga、99mWhen Tc, BFC is selected from HYNIC、MAG2、MAG3, DTPA, DOTA, TETA and NOTA.
According to the present invention, Nu is90Y or177Lu, BFC are selected from DTPA, DOTA;Nu is111When In, BFC is selected from DTPA, DOTA; Nu is64When Cu, BFC is selected from TETA, DOTA;Nu is68When Ga, BFC is selected from NOTA, DOTA;Nu is99mWhen Tc, BFC is selected from HTNIC、DTPA、MAG2、MAG3
As example, the complex formed by the polypeptide that structure of the invention is modified is as described below:
68Ga-DOTA-A-c(RGDfk);
99mTc-HYNIC-A-3PRGD2
177Lu-DOTA-A-L-3PRGD2
It should be appreciated that all isomers of the polypeptide of above structure modification of the invention, including it is enantiomter, diastereomeric Isomers and racemoid belong to the scope of the present invention.The present invention includes the stereoisomer of optical voidness form or mixture, It also include racemic mixture.Such as the amino in the structure A or L in aforementioned polypeptides exists with L- or D-shaped formula.
Those skilled in the art are known, and complex as defined above is put when bifunctional chelating agent cannot be occupied as ligand When all co-ordination positions of penetrating property nucleic, it is also necessary to synergy modes.The radionuclide and double function of synergy modes are needed in the present invention Energy chelating agent is well-known to those skilled in the art.Such as99mWhen Tc is using HYNIC as bifunctional chelating agent, match as collaboration Body, can be identical or different, be it is well known in the prior art those, wherein common synergy modes include water-soluble phosphine (such as three sulfonate sodium TPPTS of triphenylphosphine), N- tri- (methylol) methylglycine (Tricine), N- bis- (ethoxy) Glycine, gluceptate, ethylenediamine-N, N '-diacetate esters (EDDA), 3- benzoyl pyridine (BP), pyridine -2- azo - P- dimethylaniline (PADA) etc..For example,99mWhen Tc is using HYNIC as bifunctional chelating agent, TPPTS and Tricine are as collaboration Ligand.And for example177Lu or68When Ga is using DOTA as bifunctional chelating agent, does not need synergy modes and complete coordination.As reality Example, provides the complex such as flowering structure:
The present invention also provides the molecular probes with above-mentioned complex structure.
The present invention also provides the preparation methods of the rgd peptide of above structure modification, which comprises
Optional step (1): the coupling reaction of A and L;
Step (2): the coupling reaction of A- (L) n structure division and rgd peptide.
Preparation method according to the present invention, further includes the synthesis step of A, and the step includes:
(3) formula 1:With formula 2:Coupling reaction.
Preparation method according to the present invention, the coupling reaction of step (1), (2) and (3) are carboxyls and ammonia well known in the art The amidation process of base, it usually needs first activate its carboxyl, then reacted again with amino.Such as it can add first in the solution of formula 1 Enter N- hydroxy benzo triazole or n-hydroxysuccinimide (NHS) forms amide, which can be added condensing agent carbodiimide, Such as DCC (dicyclohexylcarbodiimide), DIC (N, N'- diisopropylcarbodiimide), promote the generation of amide.Subsequent amide In the presence of condensing agent, such as EDCI, HOBT, DIEA and the reaction of formula 2 obtain object, and reaction dissolvent can select dichloro Methane, DMF etc..Step (2), the reaction of step (1) are identical as step (3), are similarly first activated carboxyls, then at amide, because This can be reacted under similarity condition.
The present invention also provides the preparation method for the radio nuclide compound that the rgd peptide modified by above structure obtains, The described method includes:
Optional step (1): the coupling reaction of A and L;
Step (2): the coupling reaction of A- (L) n structure division and rgd peptide;
Step (3): the coupling reaction of A- (L) n-RGD polypeptide and bifunctional chelating agent;
Step (4): with A- (L) n-RGD polypeptide of radioisotope labeling connection bifunctional chelating agent.
According to the present invention, the preparation method of the complex, further includes the synthesis step of A, and the step includes:
Formula 1:With formula 2:Coupling reaction.
Preparation method according to the present invention, the coupling reaction of step (3) be equally amino in A- (L) n-RGD polypeptide with The amidation process of the carboxyl of bifunctional chelating agent, thus can be used with step (1), (2) identical reaction condition, such as In the presence of condensing agent, including EDCI, HOBT, DIEA, use methylene chloride, DMF etc..The radionuclide of step (4) Label be well-known to those skilled in the art, such as by radionuclide be added molecular probe precursor, that is, connect difunctional In the solution of A- (L) n-RGD polypeptide of chelating agent, heating is obtained.The heating temperature is at 80-120 DEG C, and the reaction time is in 5- 60min.Usually after label purifying and Quality Control described in marker, such as with known Sep-Pak reverse-phase chromatographic column, Sephadex G-25 column, YMC-Pack ODS-A C18 analytical column, Radio-HPLC method etc. purify and measure mark rate and radiation Chemical purity.
The present invention also provides a kind of pharmaceutical composition, the composition includes a effective amount of above-mentioned labelled coordination compound Nu- BFC-A- (L) n-RGD polypeptide.
According to the present invention, when Nu is diagnosis nucleic, pharmaceutical composition of the invention is a kind of diagnostic medicine, such as described Localization diagnosis of the drug as imaging agent, for integrin alpha v beta 3 positive tumor.It is directly applied to individual, is given by detection The radioactive ray that the compound of subject's application is emitted, the information obtained based on the radioactive ray image to diagnose.Preferably, originally The diagnostic medicine of invention is a kind of injectable formulation, and it includes the carriers of above-mentioned labelled coordination compound and injectable.It is preferred that described aobvious As agent refers to as positron emission computerized tomography PET, single photon emission computerized tomography SPECT according to the present invention, When Nu is treatment nucleic, pharmaceutical composition of the invention is a kind of therapeutic agent, and the drug is positive for integrin alpha v beta 3 The radiation targeted therapy of tumour.It is directly applied to individual, the drug and integrin alpha v beta 3 have specific affinity, rich Collection is in tumor tissues, and radionuclide is by emitting pure beta rays or generating ionising radiation biology with gamma-ray beta rays Effect destroys pathological tissues.Preferably, therapeutic agent of the invention is a kind of injectable formulation, and it includes above-mentioned labelled coordination compounds With the carrier of injectable.
Preferably, pharmaceutical composition of the invention is a kind of intravenous injection, such as a kind of colourless transparent liquid injection.It is suitable Auxiliary material for intravenous injection is it is known in the art that described pharmaceutical composition can be only fitted in aqueous solution, if it is desired, Using the buffer of PHYSIOLOGICALLY COMPATIBLE, including for example, phosphate, histidine, citrate etc. can also make for adjusting preparation pH With tonicity agent, cosolvent, such as polyethylene glycol, hypotoxicity surface-active is also can be used in sodium chloride, sucrose, glucose etc. Agent, such as polysorbate or poloxamer etc..
Preferably, pharmaceutical composition of the invention further includes anti-adsorbent, such as physiological saline, 1% cyclodextrin it is water-soluble Liquid, the PBS solution (mass fraction of such as tween is 0.01~0.1%) containing Tween-20, contains anti-adsorbent using of the invention Pharmaceutical composition can effectively avoid institute using the solution of surfactant such as Tween-20 (mass fraction such as 0.05%) State non-specific adsorption of the marker in infusion, hence it is evident that prevent absorption of the tube wall to radioactively labelled substance, realize dosage Precision, while avoiding the waste of drug in use.Pharmaceutical composition containing above-mentioned anti-adsorbent is in a system It arranges and is continuously shifted in new EP pipe 4 times, the rate of recovery of marker is up to 97% or more.
The present invention also provides above-mentioned A- (L) n-RGD polypeptides or Nu-BFC-A- (L) n-RGD peptide molecule probe to prepare Purposes in drug.According to the present invention, for diagnosing or treating integrin alpha v beta 3 positive tumor, the tumour is the drug Refer to entity tumor, such as blood, liver, body of gland (such as mammary gland, prostate, pancreas), intestines (such as colorectum), kidney, stomach, The malignant tumour at the positions such as spleen, lung, muscle, bone.
The present invention also provides a kind of diagnosis or the highly expressed hematological systems for the treatment of integrin alpha v beta 3 and solid malignant A effective amount of above-mentioned Nu-BFC-A- (L) n-RGD polypeptide is applied to the individual of this demand by method.According to the present invention, described Individual can be mammal, such as mankind.
Beneficial effect
The present invention has designed and prepared the rgd peptide of structural modification, and is prepared by the rgd peptide of the structural modification The Novel series rgd peptide molecular probe that pharmacokinetic property improves.It is a discovery of the invention that the rgd peptide passes through structure Modification and transformation, the molecule of the present invention formed by the rgd peptide of the structural modification by bifunctional chelating agent and radionuclide Probe internal external stability with higher, the Percentage bound with albumin (albumin), to be obviously prolonged half-life period;This The molecular probe of invention tumor uptake rate with higher, contrast, safety, reduce dosage, reduce side effect, So as to improve RGD plurality of probes as imaging results of the localization diagnosis molecular probe in SPECT and/or PET and conduct Treat the effect that molecular probe carries out radioactivity magnetic target therapy.
One example of novel molecular probe of the present invention177Lu-DOTA-A-L-3PRGD2, there is higher take the photograph in blood It takes, to improve the intake in tumour, 8 times or so of the reachable former probe of accumulative intake in blood are in tumour Accumulation intake be the 4 of former probe
Times or so.Its 4 hours after injection, tumor uptake reached highest, and percentage injection dose rate is 26.52 ± 0.58%ID/g remains to clearly image after 48 hours after injection.This property significantly improves RGD plurality of probes and examines as imaging The imaging results of disconnected molecular probe,
And the effect of radioactivity magnetic target therapy is carried out as treatment molecular probe.
Term definition and explanation
Unless otherwise defined, the connotation that all scientific and technical terminologies have herein and claim theme fields technology The normally understood connotation of personnel is identical.Unless otherwise indicated, all patents, patent application, the public material being cited in full text herein It is integrally incorporated by reference herein.If there are multiple definition to term herein, it is subject to the definition of this chapter.
Rgd peptide: it is known in the art substance.RGD is containing arginine-glycine-aspartic acid (Arg-Gly- Asp) the micromolecule polypeptide of amino acid sequence.D type phenylalanine and valine is added, has synthesized RGD ring pentapeptide structure-c (RGDfV), wherein c represents the polypeptide as annular, R represents arginine, G represents glycine, D represents aspartic acid, f represents D- benzene Glycine, V represent valine.5 amino acids of ring pentapeptide structure-c (RGDfV) are obtained c after other amino acid substitutions (RGDfK), c (RGDfE), c (RGDyk), wherein K is lysine, E is glutamic acid, y is D-Tyrosine.Such as c (RGDfK) tool Just like flowering structure:
These cyclic peptide structures can form dimer, such as E [c (RGDyk)]2、E[c(RGDfK)]2, with glutamic acid by two RGD cyclic peptide connects to form dimer.3PRGD2Refer to containing there are three the polyethyleneglycol modified five rings RGD peptide dimer, i.e. PEG4- [PEG4-c(RGDXk)]2, X is D-PG, D-Tyrosine etc..Illustratively, structural schematic diagram is as follows:
Bifunctional chelating agent: bifunctional chelating agent (bifunctional chelator BFC) refers to can be with biology point Son is covalently attached energy chelating metal nucleic again, and structure can guarantee the firm connection with metal nucleic, and the metal nucleic introduced Far from biomolecule to guarantee that its bioactivity is not suffered a loss, a stable nucleic-chelating agent-biomolecular labeling object is formed This kind of functional organic materials.The bifunctional chelating agent that the present invention uses is those of known in state of the art, example Such as HYNIC (buzane niacinamide), MAG2(mercaptoacetyl Diglycocol), MAG3(mercaptoacetyl triglycine), DTPA (diethyl Base pentaacetic acid), DOTA (- 1,4,7,10 tetraacethyl of Cyclen), NOTA (tri- azepine of 1,4,7- - 1,4,7 tricarboxylic acids of cyclononane), TETA (- 1,4,8,11 tetraacethyl of 1,4,8,11- tetraazacyclododecane tetradecane) etc..
The term as used herein " treatment " includes alleviating, mitigate or improving disease or illness disease with other similar synonyms Shape prevents other symptoms, improves or prevents the potential metabolism reason for leading to symptom, inhibit disease or illness, such as prevent disease Or the development of illness, alleviate disease or illness, disease or illness is made to improve, alleviates the symptom as caused by disease or illness, or Stop the symptom of disease or illness, in addition, the term includes the purpose of prevention.The term further include obtain therapeutic effect and/or Preventive effect.The therapeutic effect refers to the potential disease that healing or improvement are treated.In addition, to relevant to potential disease one The healing or improvement of kind or a variety of physiological signs are also therapeutic effect, such as although patient may nevertheless suffer from the shadow of potential disease It rings, but observes that patient profiles improve.For preventive effect, described group can be applied to the patient for suffering from specified disease risk Close object, even if not yet make medical diagnosis on disease, but apply institute to the patient for one or more physiological signs of the disease occur State composition.
Detailed description of the invention
Attached drawing 1: 1 final product of embodiment68The radioactivity HPLC of Ga-DOTA-A-c (RGDfk) schemes;Fig. 1 a is figure before purification 1b is after purification;
Attached drawing 2: the molecular probe and control probe of embodiment 1 are in lotus LLCC57BL/6J mouse vivo biodistribution distribution results;
Attached drawing 3: 3 final product of embodiment99mTc-HYNIC-A-3PRGD2Mark rate measurement chart;
Attached drawing 4: the serum of embodiment 3 removes experimental result picture;
Attached drawing 5: the lotus U87MG nude mice SPECT/CT of embodiment 3 images figure;
Attached drawing 6: attached drawing A B C D be respectively embodiment 12 SPECT/CT image results molecular probe of the present invention with it is right According to the %ID/g in U87 tumour of probe, tumour/kidney, tumour/muscle, tumour/liver ratio compares figure;
Attached drawing 7: the molecular probe of embodiment 3 distribution results figure in lotus U87MG nude mouse;
Attached drawing 8: the final product of embodiment 7177Lu-DOTA-A-L-3PRGD2Radioactivity HPLC figure;
Attached drawing 9: the molecular probe serum of embodiment 7 removes experimental result picture;
Attached drawing 10: the molecular probe lotus U87MG nude mice SPECT/CT of embodiment 7 images figure;
Attached drawing 11: the molecular probe of embodiment 7 is in U87 mouse vivo biodistribution distribution map;
Attached drawing 12: the tendency chart that the gross tumor volume of the molecular probe radiation treatment of embodiment 7 changes over time;
Attached drawing 13: bio distribution of the molecular probe of embodiment 7 in MC-38 mouse model, MC-38 nude mice SPECT/CT Imaging figure and the tendency chart that the gross tumor volume of radiation treatment changes over time in MC-38 mouse model.
Specific embodiment
General formula compound and its preparation method and application of the invention is done further below in conjunction with specific embodiment Detailed description.It should be appreciated that the following example is merely illustrative the ground description and interpretation present invention, and it is not necessarily to be construed as to this The limitation of invention protection scope.All technologies realized based on above content of the present invention are encompassed by the model the present invention is directed to protection In enclosing.Unless otherwise indicated, raw materials and reagents used in the following embodiment are commercial goods, or can pass through known formula Method preparation.
Statistical analysis
Experimental result is indicated in the form of average value ± standard deviation (mean ± SD).Difference between group uses variance Analysis and t, which are examined, carries out statistical analysis to result.P < 0.05, it is believed that have statistical difference (*).
Embodiment 168The preparation of Ga-DOTA-A-c (RGDfk)
The synthetic route chart of DOTA-A-c (RGDfk) is as follows:
(1) synthesis of compound 1
It weighs 4- (4- iodophenyl) butyric acid (9.8mg, 33.8mmol) and single port bottle is added, be dissolved in 400 μ L DMF, be added NHS (3.9mg, 33.9mmol).Add 5.3 μ L DIC.30 DEG C of constant temperature are stirred to react 2h, TLC (ethyl acetate: petroleum ether: second Acid=100:200:2) it monitors to raw material disappearance.To the end of reacting, reaction solution is dissolved in ethyl acetate, is washed with water three times, acetic acid Ethyl ester is mutually dry with anhydrous sodium sulfate, column chromatography for separation (ethyl acetate: petroleum ether: acetic acid=100:200:2) after concentration, collects Fraction detection, the product of collection is spin-dried for obtain white solid 10.2mg, yield 78%.Confirm through MALDI-TOF mass spectral analysis For expected product.
MS (ESI): m/z=387.2137 (chemical formula: C14H14INO4, calculate molecular weight 387.00).Take a small amount of solid weight After new dissolution, through HPLC Purity purity > 98%.
(2) synthesis of compound 2
It takes compound 1 (20mg, 51.7mmol) to be added in the round-bottomed flask equipped with 500 μ L DMF to dissolve, then takes Fmoc-D- Flask is added in Lys.HCL (22.8mg, 56.3mmol), and is added 12 μ L DIEA and adjusts pH=8.5.30 DEG C of constant temperature are stirred to react 0.5h, TLC (ethyl acetate: petroleum ether: acetic acid=100:200:1) monitoring to raw material disappear.After reaction, reaction solution is molten In 20mL ethyl acetate, washed 3 times with 20mL saturated sodium chloride solution, 20mL respectively.Ethyl acetate layer is dry with anhydrous magnesium sulfate Dry, vacuum rotary steam obtains yellow mucus 26.5mg to doing.Yield 85%.Expected production is confirmed as through MALDI-TOF mass spectral analysis Object.
MALDI-TOF-MS:m/z=1718.33 (M+H)+, 1741.02 (M+Na)+(chemical formula: C80H119N25O16S is calculated Molecular weight: 1719.02Da.).It after taking a small amount of product to re-dissolve, is analyzed using HPLC, purity 96.7%.
(3) synthesis of compound 3
Single port bottle is added in Weigh Compound 2 (10mg, 15.6mmol), is dissolved in 500 μ L DMF, adds NHS (1.83mg, 15.9mmol), then 4 μ L of DIC is added into single port bottle.It is stirred to react at 35 DEG C 35 minutes.When heating 5min, hair Existing suspension becomes clear.TLC (ethyl acetate: petroleum ether: acetic acid=100:200:1) monitoring to raw material disappears.After reaction, will Reaction solution is dissolved in 20mL ethyl acetate, is washed 3 times with 20mL saturated sodium chloride solution, 20mL respectively.Ethyl acetate layer is with anhydrous Magnesium sulfate is dry, chromatography (ethyl acetate: petroleum ether=1:1) after concentration, collects fraction detection, the product of collection is spin-dried for Vacuum rotary steam obtains white solid 8.02mg, yield 70% to doing.Expected product is confirmed as through MALDI-TOF mass spectral analysis.
MALDI-TOF-MS:m/z=760.2 (M+Na)+(chemical formula: C35H36IN3O7, calculate molecular weight: 737.16).
(4) synthesis of compound 4
It takes compound 3 (11.5mg, 15.6mmol) in 1mL EP pipe, is dissolved in 500 μ L DMF, be added c (RGDfk) (9.4mg, 15.6mmol) adds DIEA and adjusts pH=8.5, and 30 DEG C are reacted 2 hours.It is monitored and is reacted using HPLC.HPLC is (high Effect liquid phase chromatogram) method one: YMC-Pack ODS-A C18 semi-preparative column is equipped with using 1100 Series HPLC System of Agilent (10mm×250mm,Poresize, granular size are 5 μm), gradient elution 25 minutes, flow velocity 1mL/min, wherein flowing Phase A is H2O solution, B are acetonitrile (containing 0.05%TFA).Elution gradient is set as 100%A and 0%B when starting, at 20 minutes 0%A and 100%B, 100%A and 0%B at 25 minutes.Product appearance in 14.854min.
(5) synthesis of compound 5
125 μ L piperidines are added in single step reaction EP pipe forward, slough Fmoc protecting group, 500 μ L are added into reaction system Ether high speed centrifugation precipitating, discards ether, and obtaining product is white solid, about 9.55mg, yield 61%.Through MALDI- Expected product is confirmed as in TOF mass spectral analysis.
MALDI-TOF-MS:m/z [C43H62IN11O9]+(M+H)+, 1004.38;Measured value 1004.88.Take a small amount of solid After dissolution, HPLC identifies that purity is 94.4%.
(6) synthesis of compound 6
Weigh Compound 5 (5mg, 4.98mmol), DOTA-NHS-ester 3.79mg are dissolved in 400 μ L DMF.DIEA is added Adjust pH=8.5,30 DEG C of concussion reaction 30min.Half preparation HPLC is isolated and purified, and HPLC (high performance liquid chromatography) method two: is made With 1100 Series HPLC System of Agilent be equipped with YMC-Pack ODS-A C18 semi-preparative column (10mm × 250mm,pore Size, granular size are 5 μm), gradient elution 25 minutes, flow velocity 1mL/min, wherein mobile phase A was H2O solution, B are acetonitrile (containing 0.05%TFA).Elution gradient is set as 80%A and 20%B when starting, 50%A and 50%B at 20 minutes, at 25 minutes 80%A and 20%B.Fraction when retention time is 15.7 minutes is collected, collection liquid is merged and is lyophilized, white powder is obtained 3.2mg.Yield is 46.3%, purity > 95%.Expected product is confirmed as through MALDI-TOF mass spectral analysis.
MALDI-TOF-MS:m/z [C55H80IN15O16]+(M+H)+, 1390.50;Measured value 1390.7040.HPLC mirror Determining purity is 98%.
(7)68Preparation, purifying and the Quality Control of Ga-DOTA-A-c (RGDfk)
Quantitative DOTA-A-c (RGDfk) is accurately weighed, after being re-dissolved with water, 20 μ g/ pipe is dressed up, is placed in -80 DEG C of refrigerators It saves.When radioactive label, the compound dispensed is taken out, room temperature is melted 30 minutes.
Preparation: it is eluted using 0.05M HCl68Ge-68Ga generator obtains 12.4mCi (469.9MBq)68Ga liquid.Take 500 μL 68Ga leacheate is placed in clean EP pipe, and 12 μ L of 1.25M NaOAc solution is added, and adjusts pH value to 4.0.By this68Ga liquid (4.0mCi, 148MBq) is added to equipped in polypeptide EP pipe.Metal bath is heated to 100 DEG C, reacts 10 minutes.
Purifying: being purified with Sep-Pak C-18 pillar, activates Sep-Pak C-18 column with 10mL dehydrated alcohol first, And then 10mLH is used2O washes column.By radioactive sample by Sep-Pak C-18 column, column is washed with the physiology salt of 10mL, is removed Free68Ga finally washes column with the ethyl alcohol of 0.4mL 80%, collects radioactively labelled substance.0.22 μm of miillpore filter degerming is crossed, is used In the experiment in vivo in later period.
Quality Control: radioactive probe is placed at room temperature for after ten minutes, monitors purity using radioactivity HPLC.Using radioactivity HPLC Method measures mark rate and radiochemical purity.Using HPLC system, it is equipped with radioactivity on-line checking device and Zorbax C18 points Analysis column (4.6mm x 250mm,Pore size), gradient elution 30 minutes, flow velocity 1.0mL/min, wherein mobile phase A be H2O solution, B are acetonitrile (containing 0.05%TFA).Elution gradient is set as 100%A and 0%B when starting, at 5 minutes 95%A and 5%B, 78%A and 22%B at 30 minutes, returns to baseline gradient 100%A and 0%B for 30-35 minutes.
Before being marked, water and buffer used all use Chelex 100column to handle, and remove metal ion.68Ga-DOTA-A-c (RGDfk) is marked with one-step method, and preparation process is simple and quick, and mark rate is 85%, through C-18Sep-Pak column After purification, the top coal drawing of marker is all larger than 99%.After calibrated calculating, marker yield is shown in Fig. 1 80%.
Embodiment 2: the molecular probe vivo biodistribution distribution experiments of embodiment 1
Lotus knurl C57BL/6J mouse 36 is taken, is randomly divided into 9 groups, every group 4.Wherein four groups of tail veins are respectively injected 0.1mL68Ga-DOTA-A-c (RGDfk) (about 1.85MBq), four groups of tail vein injections68Ga-DOTA-c (RGDfk) is (about 1.85MBq), it is put to death after 0.5h, 1h, 2h, 4h take blood, dissects coring, liver, spleen, lung, kidney, intestines, stomach, bone, meat, tumor.It surveys Quality and radiocounting weigh and measure radioactivity cpm counting, the percentage injection of per gram of tissue is calculated after decay correction It measures (%ID/g).Remaining one group of tail vein injects 0.1mL simultaneously68Ga-DOTA-A-c (RGDfk) and 0.05mL c (RGDfk) are molten Liquid (0.5mg), puts to death after one hour, takes organ to weigh and measures radioactivity cpm counting, per gram of tissue is calculated after decay correction Percentage injection dosage (%ID/g).
Injection68Intake has reached 33.32 ± 5.49%ID/g, tumour in blood after Ga-DOTA-A-c (RGDfk) 0.5h It absorbs in 7.52 ± 0.99%ID/g, and in contrast,68After Ga-DOTA-c (RGDfk) 0.5h blood uptake values 1.49 ± 0.49%ID/g, the uptake values in tumour are 2.74 ± 0.73%ID/g.In subsequent time point,68Ga-DOTA-A-c (RGDfk) tumor uptake is consistently higher than68Ga-DOTA-c (RGDfk), after injecting 4h68Ga-DOTA-A-c (RGDfk) tumor uptake Value is 6.57 ± 0.89%ID/g, is683.5 times of Ga-DOTA-c (RGDfk), illustrate, novel probe can effectively with serum egg White combination improves the blood halflife of probe, effectively increases tumor uptake value (P < 0.01, n=4).The probe is through kidney generation It thanks.Biodistribution data is shown in Fig. 2.
Embodiment 3:99mTc-HYNIC-A-3PRGD2Preparation
HYNIC-A-3PRGD2Synthetic route schematic diagram is as follows:
R=3PRGD2
(1) synthesis of compound 1,2,3
The synthetic method reference implementation example 1 of compound.
(2) synthesis of compound 7
Weigh Compound 3 (5.64mg, 7.65mmol) is dissolved in 500 μ L DMF in 1mL EP pipe, and 3PRGD is added2 (15.75mg, 7.8mmol) adds DIEA and adjusts pH=8.5, and room temperature reaction is overnight.It is monitored and is reacted using HPLC.Using HPLC (high performance liquid chromatography) method one, product appearance in 13.17min.HPLC is collected, and through Mass Spectrometric Identification, is determined as being expected Product.
MALDI-TOF-MS:m/z=2680.89 (chemical formula: C123H181IN24O35, calculate molecular weight: 2681.22Da.).
(3) synthesis of compound 8
125 μ L piperidines are added in single step reaction EP pipe forward, slough Fmoc protecting group, then 500 μ are added into reaction system L ether high speed centrifugation precipitating, discards ether, and obtaining product is white solid, about 10.6mg, yield 56%.Through MALDI- Expected product is confirmed as in TOF mass spectral analysis.
MALDI-TOF-MS:m/z=2460.85 (M+H)+, 2482.87 (M+Na)+(chemical formula: C108H171IN24O33, meter Calculate molecular weight: 2459.15Da.).
After taking a small amount of solid to re-dissolve, purity analysis, purity 92.5% are carried out using HPLC.
(4) synthesis of compound 9
Weigh Compound 8 (5mg, 2.03mmol), SBZH-HYNIC 0.9mg are dissolved in 500 μ L DMF.DIEA tune is added Save pH=8.5,30 DEG C concussion reaction 10 minutes.Half preparation HPLC is isolated and purified, and HPLC (high performance liquid chromatography) method three: is used 1100 Series HPLC System of Agilent outfit YMC-Pack ODS-A C18 semi-preparative column (10mm × 250mm,pore Size, granular size are 5 μm), gradient elution 30 minutes, flow velocity 1mL/min, wherein mobile phase A was H2O solution, B are acetonitrile (containing 0.05%TFA).Elution gradient is set as 100%A and 0%B when starting, 75%A and 25%B at 5 minutes, at 25 minutes 50%A and 50%B, 25-30 minutes return baseline gradient 100%A and 0%B.Collect evaporating when retention time is 15.2 minutes Point, merge collection liquid and be lyophilized, obtains white powder 1.95mg.Yield is 34.8%, purity > 95%.Through MALDI-TOF mass spectrum It is analyzed to identify as expected product.
MALDI-TOF-MS:m/z=2763.07 (M+H)+, 2785.21 (M+Na)+(chemical formula: C121H180IN27O37S, meter Calculate molecular weight: 2762.18Da.).
It is analyzed after taking a small amount of solid to dissolve through HPLC, purity is greater than 99%.
(5)99mTc-HYNIC-A-3PRGD2Preparation and Quality Control
20 μ L HYNIC-A-3PRGD are sequentially added in EP pipe2(1mg/mL is dissolved in pure water), 100 μ L tricine solution (100mg/mL is dissolved in 25mM Succinate Buffer, pH 5.0), (it is slow that 60mg/mL is dissolved in 25mM succinate to 100 μ L TPPTS Fliud flushing, pH 5.0), 100 μ L Na99mTcO4(10mCi).After mixing, 100 DEG C of heating water bath 25min are set.Marked product is cooling Afterwards, its mark rate and radiochemically pure are analyzed with HPLC (HP1100 high performance liquid chromatography is furnished with LB-509 radioactive detector) Degree.
99mTc-HYNIC-A-3PRGD2Using non-SnCl2One-step method preparation.Radioactivity HPLC analyzes marker,99mTc-HYNIC-A-3PRGD2Retention time is 11.8min, the mark rate > 99% measured.As shown in Figure 3.
Embodiment 4: the molecular probe serum of embodiment 3 is except experiment
14 Kunming female 4-5 week old small white mouses are taken, are randomly divided into two groups, each group is injected 0.1mL respectively99mTc- HYNIC-A-3PRGD2,99mTc-HYNIC-3PRGD2(about 1.85MBq), after injection respectively at 1min, 3min, 5min, 7min, 10min, 15min, 20min, 30min, 60min, 90min, 120min take blood, and measurement radioactivity cpm is counted, after decay correction Calculate percentage injection dosage (%ID/g) of two kinds of probes in blood.
By serum except experiment, it may be seen that the 3PRGD after structure of modification2Probe, internal property and former probe Compared to being substantially change.99mTc-HYNIC-A-3PRGD2Drug quick half-life period be 4.57min;Half-life period is at a slow speed 93.32min.And99mTc-HYNIC-3PRGD2The drug quick half-life period of (non-structural modification) is 0.72min;Half-life period at a slow speed For 17.91min.Drug quick half-life period improves 6.3 times, and half-life period improves 5.2 times at a slow speed.1min is injected, it is unmodified Molecular probe blood uptake values are 17.07 ± 11.77%ID/g, and99mTc-HYNIC-A-3PRGD2 intake for 44.76 ± 11.83%ID/g is the former about 2.6 times.In 5min, the former uptake values only have 4.39 ± 2.55%ID/g, and the latter is this The intake of invention molecular probe is 33.63 ± 7.83%ID/g, is the former 7.6 times.When 10min, the former absorbs 1.99 ± 1.54%ID/g, and the latter's intake is 19.06 ± 6.51%ID/g.The former absorb after 60min, drop to 0.5%ID/g hereinafter, In 240min, uptake values are 0.34 ± 0.18%ID/g;In contrast, the intake of the latter is 2.81 ± 0.83%ID/g, is 8 times of former probe.As it can be seen that structural modification greatly extends 3PRGD2Blood residence time (P < 0.01, n=7), thus Be conducive to increase probe in the uptake values of tumor locus.Serum division result is shown in Fig. 4.
Embodiment 5: the molecular probe tumor bearing nude mice SPECT/CT imaging of embodiment 3
99mTc-HYNIC-A-3PRGD2(calling molecular probe of the present invention in the following text) is prepared by 1.2.3 method.Simultaneously according to 3PRGD2Mark Note method carries out99mTc-HYNIC-3PRGD2The preparation of (calling control probe in the following text).After carrying out radioactivity HPLC detection, physiology salt is used Water is diluted to 2mCi/100 μ L respectively, takes lotus U87MG tumor nude mice to be injected intravenously respectively, every mouse is via tail vein injection 100 μ L's99mTc-HYNIC-A-3PRGD2.The drug is respectively organized with the specificity intake in organ in mice with tumor by closing in fact It is verified, closed group mouse passes through the 1mg 3PRGD of 100 μ L of tail vein injection2Cold peptide, and inject 100 μ L's immediately99mTc-HYNIC-A-3PRGD2Every kind of medicine takes 50 μ Ci to be quantified simultaneously.Respectively 0.5,1,2,4,8,12, carry out for 24 hours As a result SPECT/CT Image Acquisition is shown in Fig. 5 lotus U87MG nude mice SPECT/CT imaging figure.
Fig. 6 A is99mTc-HYNIC-A-3PRGD2With99mTc-HYNIC-3PRGD2In the %ID/g of U87 tumour;6B,6C,6D Respectively99mTc-HYNIC-A-3PRGD2With99mTc-HYNIC-3PRGD2Tumour/kidney, tumour/muscle, tumour/liver ratio Value compares.As a result it is expressed as means ± SD, (n=3).
We have found that new probe is in each acquisition time, tumour can be imaged well, even in injection 24 hours Afterwards.This has longer blood residence time with probe after transformation, and then it is related to enhance tumor uptake.Such as Fig. 6 A quantitative analysis institute Show, removing of the molecular probe of the present invention at tumour is very slow, after injection 0.5,8, be for 24 hours respectively 11.85 ± 2.18%ID/ G, 21.17 ± 0.49%ID/g and 22.27 ± 1.64%ID/g.As shown in Figure 6B, the new spy compared with control probe, after modification Needle tumor/kidney ratio is improved, 1h after injection, and control probe tumor kidney ratio is 1.14 ± 0.26, is for 24 hours 1.21 ± 0.17 after injection; And molecular probe of the present invention accordingly absorb respectively 1.57 ± 0.26., 1.92 ± 0.17.3PRGD after transformation2Tumor/non-tumor ratio Also it is improved, as shown in Fig. 6 C, 6D.This shows that improved molecular probe compared with before transformation, there is better biology Distribution character, pharmacokinetic properties.
Embodiment 6: the molecular probe bio distribution of embodiment 3
24 lotus U87 nude mices are randomly divided into 6 groups, every group 4, are set according to 1h, 2h, 4h, 8h, 12h time point.Every group Nude mice is respectively through 20 μ Ci's of tail vein injection99mTc-HYNIC-A-3PRGD2, one group of injection 3PRGD2As Block group.And in Animal is put to death after injection, takes blood and main organs, weigh and measures radioactivity cpm counting.It is calculated after decay correction every Gram tissue percentage injection dose rate (%ID/g).
The preparation of bio distribution standard specimen: the 100 μ L marker solution for being used for biodistribution experiments is added by 100mL with syringe In volumetric flask, add water constant volume to 100mL, mix well, 1mL is accurately taken out with pipettor with gamma counter and measures activity meter Number, this counts 100 times of amplification, as standard injection dosage, prepares three Duplicate Samples, be averaged.
As shown in fig. 7, tail vein injection99mTc--HYNIC-A-3PRGD2Afterwards, with the extension of time, imaging agent is in U87 Intake in tumour is fallen after rising, the intake in the tumour at tetra- time points of 1h, 2h, 4h, 8h be respectively (22.38 ± 3.68%ID/g, 21.71 ± 3.61%ID/g, 25.96 ± 1.69%ID/g, 23.53 ± 3.40%ID/g).It absorbs highest Organ is kidney, and always in 30%ID/g or more, so radioactive probe should be by renal metabolism, this and SPECT/CT are aobvious As result is consistent.Also all compared with control probe height, reason is polypeptide structure of the invention for remaining each organ intake after injection Modification make it with blood circulation time is more long in vivo, in this way, the uptake values of each organ can all increase.
Embodiment 7:177Lu-DOTA-A-L-3PRGD2Preparation
(1) preparation of compound 2 is the same as embodiment 1
(2) preparation of compound 11
Weigh 100.0mg (250 μm of ol) compound 10,76.4mg (400 μm of ol) EDC.HCL and 46.0mg (400 μm of ol) NHS is dissolved in 5mL methylene chloride, is stirred overnight at room temperature.Mixture is separated and is purified by silica gel column chromatography, eluent be containing There is the methylene chloride of 2% methanol.Vacuum distillation removes solvent, obtains white powdery solids 85.2mg.HPLC gradient and time side Method are as follows: 0 minute 50% mobile phase A and 50% Mobile phase B;10% mobile phase A and 90% Mobile phase B at 25 minutes;At 30 minutes 50% mobile phase A and 50% Mobile phase B.It takes a small amount of product HPLC to examine purity (retention time is 17.7 minutes), then uses MALDI-TOF Mass Spectrometric Identification.
(3) preparation of compound 12
Weigh 85.2mg (147 μm of ol) Fmoc-Cys (tBu)-NHS compound 11,61.0mg (145 μm of ol) compound 2, 400 μ LDMF and 20 μ L DIEA are added, water bath sonicator forms suspension.400 μ L pure water, which are added, is completely dissolved the solid in solution, It is stirred overnight at room temperature.Mixture is separated and is purified through HPLC method, and the eluting peak that retention time is 20.9 minutes is collected. HPLC gradient and time method are as follows: 0 minute 50% mobile phase A and 50% Mobile phase B;10% mobile phase A and 90% at 25 minutes Mobile phase B;50% mobile phase A and 50% Mobile phase B at 30 minutes.Freeze-drying, obtains white powdery solids 67.2mg.It takes few Then volume production object HPLC method validation purity uses MALDI-TOF Mass Spectrometric Identification.
(4) preparation of compound 13
67.2mg (84.1 μm of ol) compound 12 is weighed, is reacted at room temperature 10 minutes after 20% piperidines-DMF dissolution is added.It is mixed It closes object to be isolated and purified through HPLC method, collects the eluting peak that retention time is 24.1 minutes.HPLC gradient and time method Are as follows: 0 minute 85% mobile phase A and 15% Mobile phase B;45% mobile phase A and 55% Mobile phase B at 25 minutes;At 30 minutes 85% mobile phase A and 15% Mobile phase B.Freeze-drying, obtains white powdery solids 27.8mg.Take a small amount of product HPLC method Purity is verified, MALDI-TOF Mass Spectrometric Identification is then used.
(5) preparation of compound 14
10.0mg (12.5 μm of ol) compound 13 is weighed, 10mg (13.1 μm of ol) DOTA-NHS is dissolved in 200 μ L DMF.It is added Insoluble matter ultrasound is dispersed as suspension by 10 μ LDIEA.200 μ L pure water, which are added, is completely dissolved the solid in solution, is stirred at room temperature Overnight.Mixture is isolated and purified by HPLC method, collects the eluting peak that retention time is 21.7 minutes.HPLC gradient and Time method are as follows: 0 minute 85% mobile phase A and 15% Mobile phase B;45% mobile phase A and 55% Mobile phase B at 25 minutes;30 85% mobile phase A and 15% Mobile phase B when minute.Freeze-drying, obtains white powdery solids 8.5mg.Product HPLC method Purity is verified, MALDI-TOF Mass Spectrometric Identification is then used.
(6) preparation of compound 15
8.5mg (8.8 μm of ol) compound 14 is weighed, 200 μ L 20%TFMSA-TFA are added and react 30 seconds, are added immediately 400 μ L DMF prevent product acidolysis.Mixture is isolated and purified through HPLC method, and collecting retention time is 26.6 and 26.9 points The eluting peak of clock.HPLC gradient and time method are as follows: 0 minute 85% mobile phase A and 15% Mobile phase B;45% stream at 25 minutes Dynamic phase A and 55% Mobile phase B;85% mobile phase A and 15% Mobile phase B at 30 minutes.Freeze-drying, obtains white powdery solids 1.2mg.Product HPLC method validation purity, then identifies it with MALDI-TOF mass spectrum.
(7)MAL-3PRGD2(L7) preparation
10mg (4.8 μm of ol) 3PRGD2,2.0mg (6.5 μm of ol) Mal-NHS is weighed, 200 μ LDMF and 10 μ LDIEA are added. Mixed liquor is stirred at room temperature overnight.Mixture is isolated and purified through HPLC method, and collecting retention time is 22.1 minutes Eluting peak.HPLC gradient and time method are as follows: 0 minute 90% mobile phase A and 10% Mobile phase B;60% mobile phase A at 25 minutes With 40% Mobile phase B;90% mobile phase A and 10% Mobile phase B at 30 minutes.Freeze-drying, obtains white powdery solids 6.7mg.A small amount of product HPLC method validation purity is taken, then it is identified with MALDI-TOF mass spectrum.
(8) preparation of compound 16
0.7mg (0.8 μm of ol) compound 15 is weighed, 1.8mg (0.8 μm of ol) MAL-3PRGD2 is added, 0.1M phosphoric acid is added Buffer (pH=7.0), room temperature concussion reaction are stayed overnight.Product is isolated and purified through HPLC method, is collected retention time and is 25.6 minutes eluting peaks.HPLC gradient and time method are as follows: 0 minute 90% mobile phase A and 10% Mobile phase B;At 25 minutes 60% mobile phase A and 40% Mobile phase B;90% mobile phase A and 10% Mobile phase B at 30 minutes.Merge product peak eluent into Row freeze-drying, obtains white powdery solids 1.2mg.A small amount of product HPLC detection purity is taken, MALDI-TOF mass spectrum is then used Identification.HPLC analyzes product purity > 98%, MALDI-TOF mass spectral results and shows m/z=3160.84.The experimental results showed that [M+ H]+be consistent with the theoretical molecular weight M=3161.36 of C137H215IN30O45S.
(9)177Lu-DOTA-A-L-3PRGD2Preparation, purifying and Quality Control
Weigh 20 μ g compounds 16, DOTA-3PRGD2Or DOTA-A-L, 200 μ L ammonium acetate buffers are then added (0.1M, pH=4.8) and 5~25mCi 177LuCl3.Mixed liquor is placed in 99 DEG C of air bath heaters and is reacted 20 minutes, from So after cooling to obtain the final product.Radiation selfdecomposition occurs for marker in order to prevent, and 200 μ L rough gentian aqueous acids (1mg/mL) are added.Label Object is able to maintain 6 hours or more and is placed at room temperature for stability.
177Lu-DOTA-A-L-3PRGD2、177Lu-3PRGD2 and177The radiochemical purity (RCP) of Lu-DOTA-A-L is examined It surveys using the Agilent HPLC-1260Infinity liquid chromatographic system and Agilent ZORBAX for having radioactive detector Extend-C18 (250x 4.6mm, 5um) chromatographic column (250x 10mm, 5 μm), flow velocity 1mL/min.Mobile phase A is that water (contains 0.05%TFA), Mobile phase B is acetonitrile (containing 0.05%TFA).Elute gradient and time method are as follows: 0~5 minute 90% mobile phase A and 10% Mobile phase B;60% mobile phase A and 40% Mobile phase B at 25 minutes;90% mobile phase A and 10% flowing at 30 minutes Phase B.
The final product177Lu-DOTA-A-L-3PRGD2Radioactivity HPLC figure see Fig. 8
Embodiment 8: the molecular probe blood distribution of embodiment 7
10 Kunming small white mouses are taken, is divided into two groups, injects 0.1mL respectively, 15 kunming mices is taken to be randomly divided into three groups of (n =5), pass through the embodiment of the present invention 14 of 100 μ L (740KBq) of tail vein injection177Lu-DOTA-A-L-3PRGD2, Yi Jizuo For control177Lu-3PRGD2 or177Lu-DOTA-A-L.It is temporally put after mouse injection from endocanthion and takes appropriate blood, weighing is simultaneously Measure radiocounting cpm.Calculate the percentage injection dose rate (%ID/g) of the per gram of tissue of radiopharmaceutical in blood.It is real It tests result and carries out nonlinear regression analysis with 7.0 software of GraphPad Prism, calculate plasma metabolism fixed double chamber bed (Two Phase decay) in quick half-life period and half-life period at a slow speed.
According to experimental result, Fig. 9 is as a result seen:177Lu-DOTA-A-L-3PRGD2Quick half-life period and half-life period point at a slow speed Not Wei 6.909min and 77.15min, and177Lu--3PRGD2Quick half-life period and at a slow speed half-life period be respectively 1.231min and 20.83min, it is seen that it is of the invention177Lu-DOTA-A-L-3PRGD2With177Lu--3PRGD2It compares, blood residence time has bright It is aobvious to extend, it was demonstrated that the present invention can extend residence time of the c (RGDfk) in blood to the structural modification of polypeptide really.This explanation The improved RGD probe of the present invention and MSA binding ability are strong, and probe can recycle in vivo the longer time with blood, make blood In radioactive probe concentration increase caused by.Probe is in the percentage injection dose rate of the per gram of tissue of blood and the integral of time (area under the curve, AUC) can indicate the function and effect of drug in blood.177Lu-DOTA-A-L-3PRGD2177Lu-- 3PRGD20~72 hour AUC (%ID/g-h) value is followed successively by 208.9,27.0 upon administration.The experimental results showed that the present invention It is novel177Lu-DOTA-A-L-3PRGD2With than177Lu--3PRGD2High 7.7 times of blood pharmacokinetics.Although177Lu- DOTA-A-L in blood most slowly fast half-life period is also longer than molecular probe of the invention, but its as molecular probe its His property, for example, delay in tumour and intake it is more far short of what is expected than molecular probe of the invention, referring specifically to following embodiments.
Embodiment 9: the molecular probe tumor bearing nude mice SPECT/CT imaging of embodiment 7
SPECT/CT imaging system (Mediso company) has 4 probes and parallel aperture collimator.U87-MG mice with tumor warp 100 μ L's (20MBq) of tail vein injection177Lu-DOTA-A-L-3PRGD2177Lu-3PRGD2Or177Lu-DOTA-A-L.Then it infuses Progress SPECT imaging in 1,4,8,12,24,48 and 72 hour after penetrating.Mouse uses 1.5% isoflurane-oxygen in videograph process It is anaesthetized, keeps the fixation of its prone position on toy bed.Closed group mouse is mixed into while injection of labelled object 1.0mgCold peptide.By SPECT image and CT image co-registration, then sketched the contours of in SPECT image in 3D imaging figure Area-of-interest (ROI), the percentage injection dose rate (%ID/cc) of the per gram of tissue of computation organization and organ.It images and fixed Measuring result includes physical half time and biological half-life, without decay corrections.The result is shown in Figure 10.
We have found that probe of the invention177Lu-DOTA-A-L-3PRGD2With pharmacokinetics appropriate, in tumour With high tumor uptake and tumour contrast, it is often more important that, the intake in tumour be significantly higher than always whole body other just Often tissue and organ.1~72 hour tumour quantitative result shows after mice with tumor injection177Lu-DOTA-A-L-3PRGD2Swollen Accumulative uptake values in tumor are 662.0ID/cc-h;177Lu-3PRGD2Accumulative uptake values in tumour are 158.7ID/cc-h ;177Accumulative uptake values of the Lu-DOTA-A-L in tumour are 266.3ID/cc-h.177Lu-DOTA-A-L-3PRGD2It is adopted each Collect time point, tumour can be imaged well, the imaging clearly at tumour even after injection 48 hours, and control probe background Height, contrast are low, are enriched at tumour few.As shown in Figure 10 quantitative analysis, uptake values of the molecular probe of the present invention at tumour Highest is removed very slow, and 4 hours after mice with tumor administration, tumor uptake reaches highest, and percentage injection dose rate is 26.52 ± 0.58%ID/g.This shows molecular probe of the invention compared with before transformation, has better biodistribution characteristics, medicine generation dynamic Mechanical characteristic.
Embodiment 10: the molecular probe vivo biodistribution distribution experiments of embodiment 7
16 U87-MG mice with tumor are taken, through 100 μ L's (0.74MBq) of tail vein injection177Lu-DOTA-A-L-3PRGD2。 1 after injection, 4,24 and 72 hours execution mouse (n=4);4 tumor-bearing mices are taken, are mixed with by 100 μ L of tail vein injection 740KBq's177Lu-DOTA-A-L-3PRGD2With the 3PRGD of 0.5mg2Cold peptide, 1 hour execution mouse after injection;Take 12 lotus knurls Mouse is randomly divided into 3 groups (n=4), through 100 μ L's (0.74MBq) of tail vein injection177Lu-DOTA-A-L-3PRGD2177Lu- 3PRGD2 or177Lu-DOTA-A-L, 4 hours execution mouse after injection.Blood and other Main Tissues and organ are taken, weigh and is surveyed Its radiocounting (cpm) is measured, the percentage injection dose rate (%ID/g) of per gram of tissue is calculated.
As shown in fig. 11a, 4 hours after injection,177Lu-DOTA-A-L-3PRGD2Tumor uptake value be 26.52 ± The tumor uptake value of 0.58%ID/g, 177Lu-3PRGD2 are 4.91 ± 0.92%ID/g,177The tumor uptake of Lu-DOTA-A-L Value is 4.80 ± 1.19%ID/g.Intake of the 177Lu-k428-3PRGD2 in tumour be significantly higher than 177Lu-3PRGD2 (P < And 177Lu-k428 (P < 0.0001) 0.0001).
As schemed shown in b, 1,4,24 and 72 hour tumor uptake value is respectively 19.93 ± 1.99% after mice with tumor administration ID/g, 28.57 ± 5.27%ID/g, 11.67 ± 2.80%ID/g and 2.66 ± 1.14%ID/g.In addition to tumour, the probe Intake highest in kidney, radioactive probe should be by renal metabolism, this and SPECT/CT image results are consistent. At 4 hours, tumour/kidney ratio was 2 times or more (28.57/13.70%ID/g).
As schemed shown in c, 1 hour tumor uptake value is 6.41 ± 1.52%ID/g after closed group administration, normal group administration 1 hour tumor uptake value is 19.93 ± 1.98%ID/g afterwards.The experimental results showed that the tumor uptake of closed group is substantially less than Unclosed group (P < 0.005), intake of the 177Lu-k428-3PRGD2 in U87-MG tumour has specificity.
Embodiment 11: the radioactivity targeted therapy of the molecular probe of embodiment 7
Take 35 U87-MG tumor-bearing mices to be randomly divided into 5 groups (n=7): the 1st group of mouse passes through 100 μ L of tail vein injection (18MBq's)177Lu-DOTA-A-L-3PRGD2, the 2nd group of mouse pass through 100 μ L's (18MBq) of tail vein injection177Lu-- 3PRGD2, the 3rd group of mouse pass through 100 μ L's (18MBq) of tail vein injection177Lu-DOTA-A-L;4th group of mouse passes through tail vein Inject 100 μ L's (9MBq)177Lu-DOTA-A-L-3PRGD2, the PBS that the 5th group of tumor-bearing mice injects 100 μ L, which is used as, to be compareed.Note It penetrates rear every 2 days monitoring gross tumor volumes and changes of weight, gross tumor volume reaches 1000mm3It is set as kind terminal.
As a result referring to attached drawing 12.Scheming A and figure C is molecular probe of the invention177Lu-DOTA-A-L-3PRGD2With compare Group177Lu--3PRGD2177Lu-DOTA-A-L is at same dose (18Mbq) and molecular probe of the present invention halves dosage The tendency chart that gross tumor volume when (9Mbq) changes over time.
Gross tumor volume after administration 14 days are as follows: 18MBq177Lu-DOTA-A-L-3PRGD2The gross tumor volume for the treatment of group is 401.3 ± 195.5mm3,9MBq177The gross tumor volume of Lu-DOTA-A-L treatment group is 691.3 ± 195.9mm3,18MBq177Lu-- 3PRGD2The gross tumor volume for the treatment of group is 1122.4 ± 189.6mm3,18MBq177The gross tumor volume of Lu-DOTA-A-L treatment group For 897.4 ± 178.0mm3, the gross tumor volume of PBS control group is 1336.3 ± 315.4mm3.Experiment on therapy the result shows that, infusing Penetrate dosage be 18MBq under conditions of,177Lu-DOTA-A-L-3PRGD2The therapeutic effect for the treatment of group is significantly better than177Lu-3PRGD2 Treatment group (p < 0.0001) and177Lu-DOTA-A-L treatment group (p < 0.05).Over the course for the treatment of, the weight of above three groups of mouse Variation and Survival outcome are in normal range (NR).Simultaneously the mouse weight variation ratio of three kinds of probes of injection normal range (NR) it It is interior, there is not significant anxious toxicity.Importantly, reducing177Lu-DOTA-A-L-3PRGD2Dosage to 9MBq, Its therapeutic effect is significantly higher than doubling dosage177Lu-3PRGD2(p<0.01).The experimental results showed that with tradition177Lu- 3PRGD2It compares,177Lu-DOTA-A-L-3PRGD2With better therapeutic effect.It is demonstrated experimentally that177Lu-DOTA-A-L- 3PRGD2The tumour growth after injecting in 10 days can be significantly inhibited after targeting radionuclide therapy in U87-MG mice with tumor.Treatment Group and control group the 10th day initial volume ratio (V/V0) are 1.4 and 8.0.
Figure B is molecular probe of the invention177Lu-DOTA-A-L-3PRGD2With control group177Lu--3PRGD2177Lu- The mouse weight that DOTA-A-L is at same dose (18Mbq) and molecular probe of the present invention is when halving dosage (9Mbq) with The tendency chart of time change.The experimental results showed that BALB/c nude mice is to 18MBq dosage177Lu-DOTA-A-L-3PRGD2 Tolerance restores normal after mouse weight and the routine blood indexes decline of administration group.Mouse after the administration of 18MBq dosage was at the 6th day Weight loss ratio it is maximum, and restore within the 14th day upon administration normal.The original body mass percentage of 6th day administration group and control group Number is 90.8 ± 3.7% and 98.7 ± 5.0%;The original body mass percentage of 14th day administration group is 100.5 ± 4.1% Hes 100.7 ± 3.4%.Compared with the control group, for molecular probe of the invention during 14 days, mouse weight variation is unobvious, it is seen that Molecular probe side effect of the invention is smaller, highly-safe.
Embodiment 12: test of the molecular probe of embodiment 7 in MC-38 mouse model
(1) the molecular probe vivo biodistribution distribution experiments of embodiment 7
As a result referring to Figure 13 A.The distribution situation of molecular probe of the invention at 1,4,24,72 hour.As can be known from the results, originally The molecular probe of invention reached at 4 hours with the extension of time, intake of the imaging agent in MC-38 tumour is fallen after rising Highest intake, percentage injection dose rate are 32.51 ± 4.95%ID/g., and be highest in all organs.As it can be seen that this hair Bright molecular probe has very high tumour/other organs intake ratio.
(2) the molecular probe MC-38 nude mice SPECT/CT imaging of embodiment 7
As a result referring to Figure 13 B.Probe of the invention can be imaged well in 1,4,24 hour acquisition time, tumour, It is imaging clearly at tumour, high almost without background, contrast.This shows that molecular probe of the invention compared with before transformation, has more Biodistribution characteristics well, pharmacokinetic properties.
(3) after the molecular probe for injecting embodiment 7, gross tumor volume and mouse weight variation
As a result referring to Figure 13 C and 13D.Figure 13 C is molecular probe of the invention177Lu-DOTA-A-L-3PRGD2In dosage When 18Mbq, 9Mbq, tendency chart that tumor size changes over time.As seen from the figure, molecular probe of the invention is in 18Mbq dosage When, to 20 days after the 12nd day, tumour is eliminated completely, and Figure 12 A of embodiment 11 is really not so.The MC-38 of the present embodiment Mouse is compared with the lotus U87 mouse of embodiment 18, the former has autoimmune function, and the latter is Immune deficient mice, therefore the former is more Close to the fuselage state of human patients, this can illustrate that molecular probe of the invention can excitation therapy person itself in treatment tumour Immunity function can efficiently inhibit and eliminate tumour so that therapeutic effect is more preferable.Figure 13 D is molecular probe of the invention in agent When measuring 18Mbq, 9Mbq, tendency chart that mouse weight changes over time.As seen from the figure, molecular probe of the invention was in 20 day phase Between, mouse weight variation is unobvious, it is seen that molecular probe side effect of the invention is smaller, highly-safe.
More than, embodiments of the present invention are illustrated.But the present invention is not limited to above embodiment.It is all Within the spirit and principles in the present invention, any modification, equivalent substitution, improvement and etc. should be included in protection scope of the present invention Within.

Claims (8)

1.A- (L) n-RGD polypeptide:
Wherein A is such as flowering structure:
L represents linking arm molecule, has the following structure:Wherein m is the integer of 1-8, such as 2-6, It is preferred that 5;
The L is by the amino reaction key chain in its carboxyl and A, for example, by marking the carboxyl and A key chain that are in L;
N is 0 or 1;
When n is 0, the rgd peptide passes through the carboxyl reaction key chain in its amino and A;
When n is 1, the rgd peptide passes through another carboxyl reaction key chain in its amino and L, such as when label in L When carboxyl is connected with A, labeled as the carboxyl and rgd peptide reaction key chain of * *;
Rgd peptide is rgd peptide selected from the following: c (RGDfV), c (RGDfK), c (RGDfE), c (RGDyk), E [c (RGDyk)]2、E[c(RGDfK)]2、3PRGD2
2. the complex of the radioisotope labeling of the rgd peptide containing claim 1 has such as undefined structure:
Nu-BFC-A- (L) n-RGD polypeptide
Wherein:
Nu is radionuclide, such as diagnostic imaging nucleic:111In、64Cu、99mTc、68Ga, or treatment nucleic:90Y、177Lu、89Sr、153Sm、188Re;
BFC is bifunctional chelating agent (bifunctional chelating agent), such as HYNIC, MAG2、MAG3、DTPA、 DOTA,NOTA,TETA;
When n is 0, the bifunctional chelating agent passes through the-NH in the carboxyl and A having in its structure2Reaction key chain;
When n is 1, the bifunctional chelating agent passes through the-NH in the carboxyl and L having in its structure2Reaction key chain.
Preferably, in the polypeptide RGD and complex of structural modification as above:
Rgd peptide is selected from: c (RGDfK), 3PRGD2
Nu is selected from:90Y、177Lu、111In、64Cu、99mTc;
When Nu is90Y、177When Lu, BFC is selected from DTPA, DOTA;When Nu is111In、64Cu、68Ga、99mWhen Tc, BFC be selected from HYNIC, MAG2、MAG3, DTPA, DOTA, TETA and NOTA.
Preferably, Nu is90Y or177Lu, BFC are selected from DTPA, DOTA;Nu is111When In, BFC is selected from DTPA, DOTA;Nu is64Cu When, BFC is selected from TETA, DOTA;Nu is68When Ga, BFC is selected from NOTA, DOTA;Nu is99mWhen Tc, BFC be selected from HTNIC, DTPA, MAG2、MAG3
3. complex according to claim 2, as described below:
68Ga-DOTA-A-c(RGDfk);
99mTc-HYNIC-A-3PRGD2
177Lu-DOTA-A-L-3PRGD2
Preferably, the complex has the following structure:
4. a kind of pharmaceutical composition, the composition includes the labelled coordination compound of any one of a effective amount of the claims 2-3.
5. pharmaceutical composition according to claim 4, when Nu is diagnosis nucleic, described pharmaceutical composition is a kind of diagnosis medicine Object, such as localization diagnosis of the drug as imaging agent, for integrin alpha v beta 3 positive tumor.
6. pharmaceutical composition according to claim 4, when Nu is treatment nucleic, described pharmaceutical composition is a kind of medicine Object, the drug are used for the radiation targeted therapy of integrin alpha v beta 3 positive tumor.
It include above-mentioned labelled coordination compound 7. being a kind of injectable formulation according to the pharmaceutical composition of any one of claim 4-6 With the carrier of injectable.It preferably, is a kind of intravenous injection;Preferably, described pharmaceutical composition contains 0.05% Tween-20 PBS solution.
8. the purposes of the complex of the polypeptide of claim 1 or any one of claim 2-3 in medicine preparation, the drug For diagnosing or treating integrin alpha v beta 3 positive tumor.The tumour refers to entity tumor, such as in blood, liver, body of gland (example Such as mammary gland, prostate, pancreas), intestines (such as colorectum), kidney, stomach, spleen, lung, muscle, bone position malignant tumour.
CN201910441556.7A 2019-05-24 2019-05-24 Nuclear medicine of RGD polypeptide with modified structure Active CN110227169B (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN201910441556.7A CN110227169B (en) 2019-05-24 2019-05-24 Nuclear medicine of RGD polypeptide with modified structure
PCT/CN2020/091861 WO2020238800A1 (en) 2019-05-24 2020-05-22 Targeted radiopharmaceuticals for tumors, and combination therapy of targeted radiotherapy thereof and immunotherapy under guidance of images
EP20815331.2A EP3906947A4 (en) 2019-05-24 2020-05-22 Targeted radiopharmaceuticals for tumors, and combination therapy of targeted radiotherapy thereof and immunotherapy under guidance of images
US17/310,604 US20230117927A1 (en) 2019-05-24 2020-05-22 Targeted Radiopharmaceutical for Tumor and Its Use in the Imaging-guided Combination Therapy of Targeted Radiotherapy and Immunotherapy

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910441556.7A CN110227169B (en) 2019-05-24 2019-05-24 Nuclear medicine of RGD polypeptide with modified structure

Publications (2)

Publication Number Publication Date
CN110227169A true CN110227169A (en) 2019-09-13
CN110227169B CN110227169B (en) 2020-07-07

Family

ID=67861073

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910441556.7A Active CN110227169B (en) 2019-05-24 2019-05-24 Nuclear medicine of RGD polypeptide with modified structure

Country Status (1)

Country Link
CN (1) CN110227169B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111840585A (en) * 2020-07-20 2020-10-30 厦门大学 Pharmaceutical composition for tumor immunotherapy
WO2020238800A1 (en) * 2019-05-24 2020-12-03 北京大学 Targeted radiopharmaceuticals for tumors, and combination therapy of targeted radiotherapy thereof and immunotherapy under guidance of images
CN113616818A (en) * 2020-05-06 2021-11-09 北京大学 Pharmaceutical composition for targeted radiation and immunization combined treatment of tumors
WO2023143502A1 (en) * 2022-01-29 2023-08-03 中国科学院生物物理研究所 FAP-α SPECIFIC RADIOPHARMACEUTICAL AND APPLICATION THEREOF
WO2024099245A1 (en) * 2022-11-07 2024-05-16 烟台蓝纳成生物技术有限公司 Rgd dimer compound, and preparation method therefor and use thereof

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101474415A (en) * 2009-01-16 2009-07-08 北京大学 RGD polypeptide radiopharmaceutical for integrin alphav beta3 positive tumor and preparation method thereof
CN101485891A (en) * 2009-02-13 2009-07-22 北京大学 Radioactive nuclide marked RGD polypeptide medicament and preparation method thereof
CN102600489A (en) * 2012-02-24 2012-07-25 南方医科大学南方医院 Peptide radioactive medicine comprising iRGD sequence
CN102626522A (en) * 2012-04-12 2012-08-08 韩彦江 Polypeptide radioactive diagnosis and treatment medicament based on chemotactic factor receptor CXCR4 polypeptide antagonist
CN104844806A (en) * 2015-05-11 2015-08-19 厦门大学 Targeted compound for nuclide labeling as well as preparation method and application thereof
WO2018045376A2 (en) * 2016-09-02 2018-03-08 Ikaria Inc. Functionally modified polypeptides and radiobiosynthesis
CN109045313A (en) * 2018-09-11 2018-12-21 北京大学 A kind of D-type polypeptide radiopharmaceutical and preparation method targeting HER2
CN109091683A (en) * 2018-08-10 2018-12-28 北京大学 A kind of ring type polypeptide radiopharmaceutical and preparation method thereof for αvβ6 Integrin targeting

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101474415A (en) * 2009-01-16 2009-07-08 北京大学 RGD polypeptide radiopharmaceutical for integrin alphav beta3 positive tumor and preparation method thereof
CN101485891A (en) * 2009-02-13 2009-07-22 北京大学 Radioactive nuclide marked RGD polypeptide medicament and preparation method thereof
CN102600489A (en) * 2012-02-24 2012-07-25 南方医科大学南方医院 Peptide radioactive medicine comprising iRGD sequence
CN102626522A (en) * 2012-04-12 2012-08-08 韩彦江 Polypeptide radioactive diagnosis and treatment medicament based on chemotactic factor receptor CXCR4 polypeptide antagonist
CN104844806A (en) * 2015-05-11 2015-08-19 厦门大学 Targeted compound for nuclide labeling as well as preparation method and application thereof
WO2018045376A2 (en) * 2016-09-02 2018-03-08 Ikaria Inc. Functionally modified polypeptides and radiobiosynthesis
CN109091683A (en) * 2018-08-10 2018-12-28 北京大学 A kind of ring type polypeptide radiopharmaceutical and preparation method thereof for αvβ6 Integrin targeting
CN109045313A (en) * 2018-09-11 2018-12-21 北京大学 A kind of D-type polypeptide radiopharmaceutical and preparation method targeting HER2

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
HAOJUN CHEN: "Novel "Add-On" Molecule Based on Evans Blue Confers Superior Pharmacokinetics and Transforms Drugs to Theranostic Agents", 《THE JOURNAL OF NUCLEAR MEDICINE》 *
HONG-BO PANG: "A free cysteine prolongs the half-life of a homing peptide and improves its tumor-penetrating activity", 《JOURNAL OF CONTROLLED RELEASE》 *
JIELING ZHENG: "Evaluation of 99mTc-3PRGD2 integrin receptor imaging in hepatocellular carcinoma tumour-bearing mice: comparison with 18F-FDG metabolic imaging", 《ANN NUCL MED》 *
JIYUN SHI: "Anti-tumor Effect of Integrin Targeted 177Lu-3PRGD2 and Combined Therapy with Endostar", 《THERANOSTICS》 *
KAI CHEN: "RGD-human serum albumin conjugates as efficient tumor targeting probes", 《MOL IMAGING》 *
KAM LEUNG: "68Ga-1,4,7,10-Tetraazacyclododecane-N,N",N"",N"""-tetraacetic acid-cyclo(Arg-Gly-Asp-D-Phe-Lys)", 《MOLECULAR IMAGING AND CONTRAST AGENT DATABASE》 *
XIAONA JIN: "Integrin Imaging with 99mTc-3PRGD2 SPECT/CT Shows High Specificity in the Diagnosis of Lymph Node Metastasis from Non-Small Cell Lung Cancer.", 《RADIOLOGY》 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020238800A1 (en) * 2019-05-24 2020-12-03 北京大学 Targeted radiopharmaceuticals for tumors, and combination therapy of targeted radiotherapy thereof and immunotherapy under guidance of images
CN113616818A (en) * 2020-05-06 2021-11-09 北京大学 Pharmaceutical composition for targeted radiation and immunization combined treatment of tumors
CN113616818B (en) * 2020-05-06 2023-04-25 北京大学 Pharmaceutical composition for targeted radiation and immunity combined treatment of tumor
CN111840585A (en) * 2020-07-20 2020-10-30 厦门大学 Pharmaceutical composition for tumor immunotherapy
WO2023143502A1 (en) * 2022-01-29 2023-08-03 中国科学院生物物理研究所 FAP-α SPECIFIC RADIOPHARMACEUTICAL AND APPLICATION THEREOF
WO2024099245A1 (en) * 2022-11-07 2024-05-16 烟台蓝纳成生物技术有限公司 Rgd dimer compound, and preparation method therefor and use thereof

Also Published As

Publication number Publication date
CN110227169B (en) 2020-07-07

Similar Documents

Publication Publication Date Title
CN110227169A (en) A kind of nuclear medicine drug of the rgd peptide of structural modification
AU2017268291B2 (en) PET-imaging immunomodulators
EP3613441A1 (en) Dual-target imaging molecular probe, preparation method therefor, and application thereof
Ferreira et al. Evaluation of novel bifunctional chelates for the development of Cu-64-based radiopharmaceuticals
CN103041412B (en) PET (Positron Emission Tomography) tracer with FSHR (Follicle-stimulating Hormone Receptor) targeting as well as preparation method and application thereof
CN108434468B (en) Radioiodinated protein binding ligand and application thereof
CN112043839A (en) Radioisotope-labeled polypeptide imaging agent targeting transferrin receptor and application thereof
WO2020238795A1 (en) Rk polypeptide radiopharmaceutical targeting her2, and preparation method therefor
MX2007010116A (en) Radiolabeled gallium complexes, methods for synthesis and use for pet imaging of egfr epxression in malignant tumors.
CN107308466A (en) With tumor vascular targeted polypeptide, molecular probe and its preparation method and application
CN114773433A (en) CD25 targeted polypeptide, molecular probe and application
Chakraborty et al. Tracer level radiochemistry to clinical dose preparation of 177Lu-labeled cyclic RGD peptide dimer
Qin et al. The development of a Glypican-3-specific binding peptide using in vivo and in vitro two-step phage display screening for the PET imaging of hepatocellular carcinoma
CN109045313B (en) D-type polypeptide radiopharmaceutical targeting HER2 and preparation method thereof
TW201511774A (en) Radiolabeled active targeting pharmaceutical composition and the use thereof
Velikyan et al. Preparation and evaluation of a 68Ga-labeled RGD-containing octapeptide for noninvasive imaging of angiogenesis: biodistribution in non-human primate
US4448762A (en) Complex of transferrin with ruthenium for medical applications
CN115286693A (en) Tumor targeting peptide aiming at carcinoembryonic antigen related cell adhesion molecule CEACAM6 and application thereof
WO2020238800A1 (en) Targeted radiopharmaceuticals for tumors, and combination therapy of targeted radiotherapy thereof and immunotherapy under guidance of images
CN115974962A (en) FAP (FAP-associated protein) targeted probe as well as preparation method and application thereof
CN107021998B (en) Positron nuclide labeled polypeptide for tumor imaging
CN107674117A (en) The preparation method for the Dimer San A Cyclopeptide derivatives cancer of pancreas molecular probes that Cu 64 is marked
CN107586321A (en) The preparation method of the mark modification Dimer San A probes of F 18
CN116655744A (en) Tumor PD-L1 targeted PET imaging agent labeling precursor, imaging agent and synthesis method and application thereof
CN118307640A (en) PET (polyethylene terephthalate) developer precursor, PET developer, and preparation method and application of PET developer precursor and PET developer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant