The present disclosure belongs to the technical field of biopharmaceutical. In particular, the present disclosure relates to a fusion protein of interleukin-2 and its use in the treatment or prevention of amyotrophic lateral sclerosis (ALS).
Amyotrophic lateral sclerosis is also called ALS, Lou Gehrig's disease, and motor neuron disease. For ALS patients, there is damage, degradation and death of the upper and lower motor neurons, progressive weakness and dystrophy of the limb, trunk, and thoracico-abdominal muscles, and even complete loss in the ability of the brain of controlling voluntary movements. ALS may be genetic, or be caused by other factors such as autoimmune mechanism, neurotrophic factor disorder, oxidative stress, mitochondrial dysfunction, excitotoxity and protein misfolding. People with ALS usually just live a few years, and have high mortality rate.
At present, ALS is mainly treated with drugs. Although the drugs can delay the progression of ALS, they cannot reverse the condition. There is no radical cure of ALS. Riluzole, a glutamate antagonist, has been approved by US FDA for the treatment of ALS with twice daily (50 mg per 12 hours). It can extend life by about 4 months. However, 10% of patients have liver injury, and cannot repair the existing damages in motor neurons. Edaravone, a brain protectant for scavenging free radicals, can effectively prevent the early progression of ALS and delay the deterioration of lung function. But it cannot reduce the mortality rate of the disease, nor does it delay the decline in neurological function. The edaravone is administrated twice a day, each of which is 30 mg diluted in an appropriate amount of saline and is injected intravenously within 30 minutes. Thus, frequent intravenous infusion is required during the therapeutic process, which is labor-intensive, costly and time-consuming, and has poor compliance of patients.
There is an urgent need for a drug for treating or preventing amyotrophic lateral sclerosis with a long administration interval.
The purpose of the disclosure is to provide a fusion protein of interleukin 2 and its use in the treatment or prevention of amyotrophic lateral sclerosis (ALS), with prolonged administration interval, alleviated pain of patients, reduced cost, and improved compliance and life quality of patients.
According to the first aspect of the present disclosure, a fusion protein of interleukin-2 is provided. The fusion protein comprises a human interleukin 2 or its variant, and a human serum albumin or its variant.
The human interleukin 2 or its variants may include: an amino acid sequence as shown in SEQ ID NO: 1; or, an amino acid sequence having at least 90% sequence identity with the amino acid sequence shown in SEQ ID NO:1.
The human serum albumin or its variants may include: an amino acid sequence as shown in SEQ ID NO:2; or, an amino acid sequence having at least 90% sequence identity with the amino acid sequence as shown in SEQ ID NO:2.
In the present disclosure, the amino acid sequence as shown in SEQ ID NO:1 is human interleukin 2 with 133 amino acid residues, and the amino acid sequence as shown in SEQ ID NO:2 is human serum albumin with 585 amino acid residues.
In some embodiments, the fusion protein of the interleukin 2 may include the amino acid sequence as shown in SEQ ID NO: 1 and the amino acid sequence as shown in SEQ ID NO:2.
In some embodiments, the amino acid at position 125 of the amino acid sequence as shown in the SEQ ID NO: 1 is not cysteine. The human interleukin 2 (IL-2) has an intrachain disulfide bond. When the amino acid at position 125 of the mature protein is cysteine, it may tend to form mismatched disulfide bonds with the other two cysteines, resulting in the loss of the activity of IL-2.
In some embodiments, the amino acid at position 125 of the amino acid sequence as shown in SEQ ID NO:1 may be replaced with serine or alanine. The mismatched disulfide bonds can be avoided through the mutation of the amino acid residue at position 125 of human interleukin 2 to serine or alanine, allowing the human interleukin 2 to remain active.
In some embodiments, the human interleukin 2 or its variant may be linked to the human serum albumin or its variant directly or via a linking peptide.
In some embodiments, the linking peptide is provided between the human interleukin 2 or its variant and the human serum albumin or its variant. There may be a large interval between the two units contained in the fusion protein of interleukin 2, so that the human interleukin 2 has the maximum probability of binding to the interleukin 2 receptor.
In some embodiments, the linking peptide may have a general formula of (GnS)m, in which n or m is an integer selected from 1 to 10.
In some embodiments, the linking peptide may have a general formula of (GnS)m, in which n is an integer selected from 1 to 4 and m is an integer selected from 0 to 3.
In some embodiments, the linking peptide may have a general formula of (GnS)m, in which n is an integer selected from 1-4 and m is an integer selected from 1-3.
In some embodiments, the linking peptide may have an amino acid sequence of GGGGSGGGGS.
In some embodiments, the linking peptide may have an amino acid sequence of GGGGS.
In some embodiments, the fusion protein may have a signal peptide at the N-terminal thereof. The addition of the signal peptide to the N-terminal of the fusion protein can further increase the expression level of the fusion protein.
In some embodiments, the signal peptide may be derived from the signal peptide of CD33 for secretion.
In some embodiments, the nucleic acid sequence encoding the signal peptide may be shown as SEQ ID NO:5.
In some embodiments, the fusion protein may have an amino acid sequence as shown in SEQ ID NO: 4.
In some embodiments, the fusion proteins may comprise: from N-terminal, human interleukin 2-linking peptide-human serum albumin, or human serum albumin-linking peptide-human interleukin 2.
In some embodiments, the amino acid sequence of SEQ ID NO:4 may be a protein having 728 amino acid residues, of which amino acids 1 to 133 are the human interleukin 2, amino acids 134 to 143 are the linking peptide GGGGSGGGGS, and amino acids 144 to 728 are the human serum albumin.
According to the second aspect of the present disclosure, an isolated nucleic acid molecule is provided. The nucleic acid molecule may encode the fusion protein provided in the first aspect of the present disclosure.
In some embodiments, the fusion protein may be encoded by the nucleotide sequence as shown in SEQ ID NO:3.
In the present disclosure, the codon optimization may be performed on the nucleotide sequences of the human interleukin 2 and human serum albumin disclosed on Genbank, according to the mammalian codon preference. The optimized nucleotide sequence of the fusion protein may be inserted in an expression vector, which is then transfected into a host cell for expression. The target protein may be purified, i.e., the fusion protein of interleukin 2.
The expression system of the fusion protein having the interleukin-2 and human serum albumin, can be transfected (for example, electrotransfer) into CHO cells with a plasmid carrying the fusion gene encoding the interleukin-2 and human serum albumin. Unexpectedly, a CHO monoclonal cell line which can stably and efficiently express the human recombinant protein, is obtained. Further, the monoclonal cell line of the present disclosure can express and secrete the fusion protein having the interleukin 2 and human serum albumin (i.e., the fusion protein of interleukin 2). It can greatly prolong the plasma half-life of human interleukin 2, and thus can be used for preparing the medicament associated with the expression of human interleukin 2.
In some embodiments, the expression vector may be selected from pEE14.4, pcDNA3.1 and pEE6.4, and preferably be pEE14.4.
According to the third aspect of the present disclosure, an expression system is provided. The expression system may comprise a CHO cell. The CHO cell may contain the nucleic acid molecule provided in the second aspect of the present disclosure.
The expression system may express the fusion protein provided in the first aspect of the present disclosure.
The present disclosure unexpectedly finds that the CHO cell, especially the CHO-K1 cell line, has a high expression level of the fusion protein, up to 4 g/L. The expressed fusion protein of interleukin 2 is highly active, and can strongly promote, in vitro, the proliferation of specific cells such as NK, T and/or Treg cells.
In some embodiments, the expression system may be a CHO-K1 cell, which is deposited in China General Microbiological Culture Collection Center on May 26, 2022, under CGMCC No. 45173.
According to the fourth aspect of the present disclosure, a pharmaceutical composition is provided. The pharmaceutical composition may comprise a fusion protein provided in the first aspect of the present disclosure, and a pharmaceutically acceptable carrier.
In some embodiments, the pharmaceutical composition may be in a dosage form of injection, tablet, or capsule.
In some embodiments, the pharmaceutical composition may be in a dosage form of solution for injection or lyophilized powder for injection.
In some embodiments, the pharmaceutical composition may include a pharmaceutically acceptable carrier.
In some embodiments, the carrier may be selected from excipient, diluent, filler, binder, wetting agent, disintegrant, absorption enhancer, surfactant, adsorption support, and/or stabilizer.
According to the fifth aspect of the present disclosure, provided is a method for treating or preventing amyotrophic lateral sclerosis (ALS). The method may comprise administrating to a subject of an effective amount of the fusion protein provided by the first aspect of the present disclosure, or the pharmaceutical composition provided by the fourth aspect of the present disclosure.
In some embodiments, the fusion protein or the pharmaceutical composition may be administered by injection.
In some embodiments, the fusion protein or the pharmaceutical composition may be administered by subcutaneous or intravenous infusion.
In some embodiments, the fusion protein or the pharmaceutical composition may be administered by subcutaneous injection.
In some embodiments, the fusion protein may be administered at 3×104IU to 1×106IU each dose.
In some embodiments, the pharmaceutical composition may be administered at 3×104IU to 1×106IU each dose, as measured by the fusion protein therein.
In some embodiments, the fusion protein may be administered every 7-28 days.
In some embodiments, the fusion protein may be administered every 14-28 days.
In some embodiments, the pharmaceutical composition may be administered every 7-28 days.
In some embodiments, the pharmaceutical composition may be administered every 14-28 days.
According to the sixth aspect of the present disclosure, provided is use of the fusion protein provided in the first aspect of the present disclosure in the treatment or prevention of amyotrophic lateral sclerosis (ALS).
In some embodiments, the fusion protein may be used for the prevention or treatment of the decrease in limb strength, body weight, and/or neurological function caused by ALS.
According to the seventh aspect of the present disclosure, provided is use of the pharmaceutical composition provided in the fourth aspect of the present disclosure in the treatment or prevention of amyotrophic lateral sclerosis (ALS).
In some embodiments, the pharmaceutical composition may be used for the prevention or treatment of the decrease in limb strength, body weight, and/or neurological function caused by ALS.
According to the eighth aspect of the present disclosure, provided is use of the fusion protein provided in the first aspect of the present disclosure in the preparation of a medicament for treating or preventing amyotrophic lateral sclerosis (ALS).
In some embodiments, provided is use of the fusion protein in the preparation of a medicament for preventing or treating the decrease in limb strength, body weight, and/or neurological function caused by ALS.
According to the ninth aspect of the present disclosure, provided is use of the pharmaceutical composition provided in the fourth aspect of the present disclosure in the preparation of a medicament for treating or preventing amyotrophic lateral sclerosis (ALS).
In some embodiments, provided is use of the pharmaceutical composition in the preparation of a medicament for preventing or treating the decrease in limb strength, body weight, and/or neurological function caused by ALS.
The fusion protein of interleukin-2 of the present disclosure has high biological activity and long plasma half-life.
The fusion protein of interleukin 2 of the present disclosure is capable of treating or preventing amyotrophic lateral sclerosis.
The fusion protein of interleukin 2 of the present disclosure is capable of increasing limb strength in ALS patients.
The fusion protein of interleukin 2 of the present disclosure can slow down the loss in body weight of ALS patients.
The fusion protein of interleukin 2 of the present disclosure can improve the neurological function of ALS patients.
The fusion protein of interleukin 2 of the present disclosure can improve the survival rate and prolong the survival cycle of ALS patients.
The fusion protein of interleukin 2 of the present disclosure has an excellent therapeutic effect, e.g. in improving the survival rate, survival cycle, motor nerve function, and weight loss of ALS patients.
Moreover, the fusion protein of interleukin 2 provided by the present disclosure has slowed-down hydrolysis in patient, significantly prolonged interval time of the administration, enhanced therapeutic effect, reduced total administration dosage. Correspondingly, the fusion protein of the present disclosure can relieve the patient's pain, reduce treatment cost, reduce the occurrence of adverse reactions, thereby improving the compliance and life quality of the patients. Therefore, the present disclosure brings hope to the recovery of ALS patients.
The invention is further illustrated through the examples below. It should be understood that the examples of the present invention are only intended to illustrate the present invention but not to limit it. Any simple improvement made to the present invention under the concept of the present invention is within the scope of protection claimed by the present invention.
The term “fusion protein” as used herein means a bioactive polypeptide (usually TCR or an antibody) and an effector molecule (usually a protein or peptide sequence) which are covalently linked by any appropriate method such as recombinant, chemical or other methods. If desired, one or more molecules may be fused via a linking peptide. Alternatively, the linking peptide may be used to assist in the construction of the fusion molecule. The fusion molecule is particularly preferred to be a fusion protein. In general, the fusion molecules may also include the conjugated molecules.
The terms “expression vector” and “expression construct” as used herein can be used interchangeably. When the above isolated nucleic acid molecules are connected to the vector, the nucleic acid sequence can be directly or indirectly connected to the regulatory elements on the vector, as long as these regulatory elements can regulate the translation and expression of the nucleic acid molecules. These regulatory elements can derived directly from the vector itself, or can be exogenous, i.e., be not derived from the vector itself. That is, the nucleic acid molecule is operationally connected to the regulatory element(s). In this disclosure, “operationally connected” means that a foreign gene is connected to the vector so that the regulatory element(s) in the vector (e.g. transcription- and translation-regulatory sequences) can play the intended functions of regulating the transcription and translation of the foreign gene. Of course, the polynucleotides used for encoding the heavy and light chains of an antibody can be inserted independently into different vectors, often into the same vector. Common vectors may include, for example, plasmids, bacteriophages, and the like.
Compared to a sequence, a variant having “at least 90% sequence identity” with the sequence as used herein may include an amino acid sequence having at least 90%, at least 91%, at least 92%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% sequence identity with the sequence, and having identical or similar function to that of the sequence.
The term “pharmaceutically acceptable carrier” as used herein may include any physiologically compatible solvent, dispersion medium, coating, antibacterial agent, antifungal agent, isotonic agent, delayed absorber, and the like. Particular examples can include one or more selected from, e.g. water, saline water, phosphate buffered saline (PBS), glucose, glycerol, ethanol, and their combinations. In many cases, the pharmaceutically acceptable carrier may include isotonic agents, such as sugars, polyols (e.g., mannitol, sorbitol), or sodium chloride. The pharmaceutically acceptable carrier may also include a trace amount of auxiliary substances, such as wetting agents, emulsifiers, preservatives or buffers, to extend the shelf life or potency of the protein.
The term “subject” as used herein refers to subjects who have ALS or are at risk of developing ALS. Subjects can include animals, preferably be mammals, and more preferably be humans.
The term “treatment” as used herein includes playing a role in the specific disease, disorder or condition, reducing the severity of the disease, disorder or condition, or delaying or slowing the progression of the disease, disorder or condition.
The term “prevention” as used herein means the likelihood of at least reducing the risk (or susceptibility) to developing a disease or condition (i.e., stopping at least one clinical symptom of the disease that has not developed in a patient who may be exposed or susceptible to the disease but has not experienced or showed the symptoms of the disease).
The term “effective quantity” as used herein means an amount of the therapeutic, prophylactic and/or diagnostic agent which is sufficient for the treatment, relievement, improvement, reduction of the symptoms, prevention, suppression of the progression, reduction of the severity, and/or reduction of the incidence of a disease, condition, and/or symptom when administered to a subject having or being susceptible to the disease, condition, and/or symptom.
The term “dosing” or “dosage” as used herein means an amount that can relieve or delay the progression of a disease, or degenerative or traumatic conditions. The dosage can be determined depending on the specific disease to be treated and other factors, including age, weight, health status, severity of the disease, administration route, frequency of treatment, and whether other medications are accompanied during treatment.
The term “amyotrophic lateral sclerosis” or “ALS” as used herein refers to a kind of motor neuron disease, involving the upper motor neurons (e.g., brain, brain stem, spinal cord), and the lower motor neurons (e.g. cranial nerve nucleus, spinal cord anterior horn cells) and their innervated trunk, limbs and head-facial muscles. ALS is clinically manifested as mixed paralysis with injuries of the upper and lower motor neurons. ALS is substantially classified into familial- and sporadic ALS.
The solutions of the present invention will be illustrated in combination with the examples below. Those skilled in the art should understand that the following examples are used only to illustrate the invention and not be deemed as limiting the scope of the invention. In the following, the description of the commonly known techniques is omitted to avoid unnecessarily confusing the concept of this disclosure. Such techniques are described in many publications, such as Molecular Cloning: A Laboratory manual (Fourth Edition) (Cold Spring Harbor Laboratory Press).
The examples, which are not indicated with the specific technology or conditions, are performed through the technology or conditions described in the literatures in the field or according to the product specifications. The reagents or instruments used, which are not indicated with the manufacturers, are conventional products that can be available on the market.
The inventors found the decrease of the regulatory T (Treg) cells in ALS patients is associated with the increased severity of the disease, and can predict the progression of the disease and the survival rate. It suggests that they may be a potential therapeutic target. In addition, the production, activation, and survival of the Treg cells are completely dependent on cytokine interleukin 2 (IL-2). Based on this, the inventors carried out the following experiments.
Illustrations of the terms and expressions involved in the examples are given below.
IL-2-HSA fusion protein is an abbreviated form of the fusion protein of interleukin 2.
IL-2 refers to human interleukin 2.
HSA refers to human serum albumin.
125Ala IL-2 refers to a mutant of interleukin-2 having an amino acid sequence in which the amino acid at position 125 is alanine. See the amino acid sequence shown by SEQ ID NO:1 for details.
The trade name of commercially available short-acting IL-2 is Xinjier and is manufactured by Beijing SL Pharm Co., LTD.
Edaravone is manufactured by Guorui Pharmaceutical Co., LTD, Sinopharm Group.
On the day before transfection, the density of CHO-K1 cells was adjusted to 0.5×106 cells/mL. On the day of transfection, high concentration endotoxin-free plasmids that had undergone linear treatment were prepared, the cell density and viability of CHO-K1 cells were measured, ensuring that the cell viability was greater than 97%. After the CHO-K1 cells were washed twice with CD CHO medium, an electroporation transfection reaction system was prepared: 700 μL of cell suspension and 40 μg of plasmid were mixed well and transferred into a 4 mm of electrode cup. The electrode cup was placed into an electroporator and the shock parameters were set to 300 V, 1000 μF. After one electric shock, the shocked cell suspension was transfer into preheated fresh CD CHO medium, and incubated at 37° C. for 20 minutes. The incubated cell suspension was inoculated evenly into a 96-well plate. 24 hours after the transfection, the cells were pressurized, added with CD CHO medium containing methionine sulfoximine (MSX) with the final concentration of 25-50 μM, and statically cultured under 5% CO2, at 37° C.
After growing to an appropriate size in the 96-well plate, the monoclonal cells were selected and transferred to a new 96-well plate. The selected monoclonal cells were incubated in 5% CO2 at 37° C. for static culture. After the cells in the well grew to confluency, the supernatant was taken from the well plate for reducing electrophoresis to detect the expression of the fusion proteins. Nine clones with the highest expression levels were selected (see
On the day (recorded as DO) of starting the culture for expression, IL-2-HSA/CHOK1 cells in 25 mL of basal medium containing 25˜50 μM MSX were inoculated into a 125 mL of shake flask at a density of 0.3×106 cells/mL, and cultured with 5% CO2 at 37° C. on a shaker at 135 rpm. On D4 of inoculation, the samples were taken and counted, and the culture temperature was decreased to 33° C. when the cell density reached to 10×106 cells/mL. On D5, fed-batch culture was carried out and the glucose concentration was controlled to 3˜4 g/L. On D13 of culture, the culture was terminated, the cell culture supernatant was harvested. Then, the expression level of the fusion protein was measured to be 4.36 mg/mL.
A culture kinetics curve of IL-2-HSA/CHOK1 cells is shown in
A kinetics curve of the expression level of IL-2-HSA/CHOK1 cell supernatant is shown in
A non-reducing electrophoresis analysis of the protein expression of IL-2-HSA/CHOK1 cell supernatant is shown in
IL-2-HSA/CHOK1 cells were inoculated into a 50 mL of shaking flask at a density of 2×106 cells/mL, and cultured with 5% CO2 at 37° C. on a shaker at 135 rpm. The samples were taken and counted daily to observe cell status, and the basal medium containing 25˜50 μM MSX was supplemented. The cell density was daily adjusted to 2×106 cells/mL until the volume of the cell culture solution reached to 300 mL. Then, stop supplement the basal medium and continue the culture, at which point it was recorded as DO. The samples were taken and counted daily and 1 mL of culture supernatant was retained. The culture temperature was decreased to 33° C. when the cell density reached to 6×106˜7×106 cells/mL. On D2, the fed-batch culture was started and the glucose concentration was controlled to 3 g/L. On D10 of culture, the culture was terminated, the cell culture supernatants were harvested, and the protein expression levels of the supernatants were measured from DO to D10. The expression level of fusion protein in the supernatant harvested on D10 was measured to be 3.12 mg/mL.
A kinetics curve of IL-2-HSA/CHOK1 cell culture is shown in
A kinetics curve of the expression level of IL-2-HSA/CHOK1 cell supernatant is shown in
A non-reducing electrophoresis analysis of the protein expression of IL-2-HSA/CHOK1 cell supernatant is shown in
Compared with the fed-batch culture in shake flasks in the prior art, the cell density and cell survival rate of the present disclosure are very high, as shown in
In this example, NK-92 cells (ATCC® CRL-2407 ™, an IL-2 dependent NK cell line derived from the peripheral blood monocytes of a 50 year-old male patient with malignant non-Hodgkin's lymphoma) were utilized to evaluate the biological activity of IL-2-HSA.
It can be seen from
In this example, CTLL-2 cells (ATCC® TIB™, a mouse cytotoxic T lymphocyte cell line, IL-2 dependent) were utilized to evaluate the biological activity of IL-2-HSA.
Referring to the human interleukin-2 biological activity assay (CTLL-2 cell/MTT colorimetric assay) in the Chinese Pharmacopoeia, CTLL-2 cells were inoculated into 96-well plate at a density of 30000 cells/well, and added with a series of gradient diluted national standards and IL-2-HSA, and cultured in 5% CO2 at 37° C. for 18 to 24 hours. Then, MTS reagent was added and incubated for 1 to 4 hours. After shaking and mixing well, the absorbance was measured at a wavelength of 490 nm using a microplate reader. The data was analyzed using GraphPad Prism 8 software, with the logarithm of dilution X as the horizontal coordinate and OD490 as the vertical coordinate. The drug action curve was fitted with four parameters. The EC50 values obtained are shown in Table 2, and the proliferation curve is shown in
The biological activity of IL-2-HSA is calculated using the following formula:
After calculation, the specific activity of IL-2-HSA was 8.38×106 IU/mg, comparable to the specific activity (not less than 1×10−7 IU/mg) of IL-2 as specified in the pharmacopoeia. However, the difference between the molecular weights of IL-2-HAS and IL-2 was about 5 times. Thus, the specific activity of IL-2-HSA was higher.
This trial included 19 animals. G1 group: 4 wild-type C57BL/6 mice, as the wild-type control group. 15 B6SJL-Tg (SOD1-G93A) mice were randomly divided into 3 groups by body weights, respectively: G2 group: model control group; G3 group: Edaravone group; G4 group: VDJ010 group, i.e. IL-2-HSA fusion protein group. Among them, G 2 group: 4 mice, G3 group: 5 mice; G4 group: 6 mice. The specific information of the animal grouping and dosing regimen is shown in Table 3.
Administration: ALS transgenic model of B6SJL-Tg (SOD 1-G93A) transgenic mice were treated on day 90 after birth; Edaravone was administrated once a day; and the IL-2-HSA fusion protein was administrated once a week.
Weight: weighing in the morning, twice a week. Results are shown in
Rotarod test: the mice of the control group and the treatment group simultaneously started to rotate the rod twice a week from day 90. One week prior to recording experimental data, the mice were trained to adapt and learn the rotating of the rod, so that they could keep rotating the rod for 3 minutes (35 rpm/min). The rotarod instrument was operated to have acceleration of 35, slowly reach 35 rpm and maintained at this speed. Each mouse was measured three times for 300 seconds with an interval of 30 min. The three rotation times were averaged for statistics. The results are shown in
Neurological scoring: From day 90, the mice from G1 to G4 groups were scored for motor neurological function twice a week. This scoring criteria was based on the 0 to 4 evaluation system recommended by ALS therapy development institute (ALS TDI). For a score of 0: the hindlimbs of the mouse can fully splay during tail suspension, and can hold for 2 sec. For a score of 1: the hindlimbs of a mouse cannot fully splay or cannot splay during tail suspension, or tremble. For a score of 2: the toes of the mouse curl downwards at least twice at least twice during a 30 cm (12 inches), or any part of the foots is dragging along on the table. For a score of 3: there is severe paralysis or minimal joint movement, only the forelimbs are used for forward crawling. For a score of 4: the mouse can not turn over within 30 sec. The results are shown in
Observation of the survival period of the experimental animals: the experimental animals were observed for their physical state until the end of the experiments. The death time of each animal was recorded. The survival rates and the survival cycles of the mice were also calculated. The results are shown in
The interaction between the IL-2-HSA fusion protein and IL-2 receptors (IL-2Rs) of different species (human, dog, rat, mouse) were analyzed by Bio-Layer Interferometry (BLI). IL-2Rs of different species were captured through ProA or hFc probes. The IL-2-HSA fusion protein was diluted to different concentration gradients. Then, the association and dissociation of the IL-2-HSA fusion protein with the IL-2Rs of different species attached to ProA or hFc probes were measured, to determine the affinity of the IL-2-HSA fusion protein with the IL-2Rs of different species. The specific processes are described as follows:
The binding abilities of the IL-2-HSA fusion protein with the IL-2Rs of different species are shown in
The above results indicate that: 1) the IL-2-HSA fusion protein binds to all the IL-2Rs of human, rat, mouse and dog, and has cross-species property; 2) the affinity of the IL-2-HSA fusion protein with the IL-2Rs of different species is ranked as follows: human>dog>rat>mouse.
In this example, the effect on the in vitro proliferation of Treg (CD3+CD4+CD25+CD127low/−) subpopulation from peripheral blood mononuclear cells (PBMCs) of healthy people of the IL-2-HSA fusion protein was compared with that of commercial rhIL-2 (R&D, Cat. No.: 202-IL) by using flow cytometry.
1. Extraction of PBMC from Human Blood
Fresh anticoagulant blood was collected from healthy people, and PBMCs were extracted with lymphocyte isolation solution (Ficoll-Paque PREMIUM, Cytiva, Cat. No.: 17-5442-02). 3-5 mL of lymphocyte isolation solution was added to a 15 mL centrifuge tube, the diluted blood sample (3 mL peripheral blood and 3 mL PBS mixed) was carefully added to the upper layer of the lymphocyte isolation solution, centrifuged at 400 g for 30-40 min. The layer of white blood cells were removed, washed twice with PBS, resuspended in RPMI-1640 medium+10% FBS and counted.
The effects of the IL-2-HSA fusion protein and rhIL-2 on the in vitro proliferation of Treg (CD3+CD4+CD25+CD127low/−) subpopulation in human PBMC are shown in
According to the results as shown in
In this example, the pharmacokinetics (PK) of the IL-2-HSA fusion protein was investigated in SD rats and Beagle dogs.
(1) Pharmacokinetics of the IL-2-HSA Fusion Protein after a Single Subcutaneous and Intravenous Administration in SD Rats.
The trial included 40 SD rats, which had no administration history, and were randomly divided into 4 groups of 10 rats each, half male and half female. The animals in Groups 1-3 were administered with a single subcutaneous injection on the nape of neck. The low-, medium- and high-dosing groups were administered with 5×105 IU/kg, 1×106 IU/kg, 2×106 IU/kg, respectively. The animals in Group 4 were administered with tail intravenous injection at 1×106 IU/kg. All of the volumes of administration were 2 mL/kg. PK blood samples were collected from the test animals at the following time points:
The concentrations of IL-2-HSA in the serum of SD rats were quantified by validated ELISA test. The pharmacokinetic parameters were calculated using noncompartmental analysis of WinNonlin software (Phoenix™, Version 8.1).
The major pharmacokinetic parameters are shown in Table 5, after the SD rats were administered with a single subcutaneous injection of different dosages (5×105, 1×106, 2×106 IU/kg) of and a single intravenous injection of 1×106 IU/kg of the IL-2-HSA fusion protein.
(2) Pharmacokinetics of the IL-2-HSA Fusion Protein after a Single Subcutaneous and Intravenous Injection in Beagle Dogs.
The trial included 24 Beagle dogs, which had no administration history, and were randomly divided into 4 groups of 6 dogs each, half male and half female. The animals in Groups 1-3 were administered with a single subcutaneous injection on the nape of neck. The low-, medium- and high-dosing group administered with 3×105 IU/kg, 6×105 IU/kg, 1.2×106 IU/kg, respectively. All of the volumes of administration were 0.6 mL/kg. The animals in Group 4 were administered with intravenous injection at 6×105 IU/kg. PK blood samples were collected from the test animals at the 30 following time points:
The concentrations of IL-2-HSA in the serum of Beagle dogs were quantified by validated ELISA test. The pharmacokinetic parameters were calculated using noncompartmental analysis of WinNonlin software (Phoenix™, Version 8.1).
The major pharmacokinetic parameters are shown in Table 6, after the Beagle dogs were administered with a single subcutaneous injection of different dosages (3×105, 6×105, 1.2×106IU/kg) of and a single intravenous injection of 6×105 IU/kg of the IL-2-HSA fusion protein.
Pharmacokinetic studies have shown that the half-life of IL-2-HSA in rats and dogs is significantly longer compared with that of natural IL-2 (several minutes in the human body). Thus, the frequency of administration and the total administration dosage can be reduced, the adverse reactions and treatment costs can be decreased, thereby improving the compliance and quality of life of patients.
The above describes the preferred embodiments of the invention in detail. However, the invention is not limited to the specific embodiments. Within the scope of the conception of the invention, various simple modifications to the technical solutions of the invention can be made, and such modifications are also within the scope of protection of the invention.
It should also be indicated that, without contradiction, the specific technical features described in the above specific embodiments can be combined in any appropriate way. To avoid unnecessary duplication, the invention does not further illustrate any possible combinations.
In addition, various embodiments of the invention may also be combined arbitrarily, provided that they do not contravene the conception of the invention. They shall also be regarded as the contents disclosed by the invention.
Number | Date | Country | Kind |
---|---|---|---|
202110982310.8 | Aug 2021 | CN | national |
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/CN2022/106115 | 7/15/2022 | WO |