CN115944741A - Application of fusion protein containing TGF-beta receptor and taxus and nucleoside analogue in preparing medicine for treating pancreatic cancer - Google Patents

Application of fusion protein containing TGF-beta receptor and taxus and nucleoside analogue in preparing medicine for treating pancreatic cancer Download PDF

Info

Publication number
CN115944741A
CN115944741A CN202211173875.2A CN202211173875A CN115944741A CN 115944741 A CN115944741 A CN 115944741A CN 202211173875 A CN202211173875 A CN 202211173875A CN 115944741 A CN115944741 A CN 115944741A
Authority
CN
China
Prior art keywords
tgf
fusion protein
weeks
receptor
administered
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202211173875.2A
Other languages
Chinese (zh)
Inventor
邹建军
张晓静
王琳娜
夏叶
黄毅慧
严平
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.)
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
Original Assignee
Jiangsu Hengrui Medicine Co Ltd
Shanghai Hengrui Pharmaceutical Co Ltd
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 Jiangsu Hengrui Medicine Co Ltd, Shanghai Hengrui Pharmaceutical Co Ltd filed Critical Jiangsu Hengrui Medicine Co Ltd
Publication of CN115944741A publication Critical patent/CN115944741A/en
Pending legal-status Critical Current

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The disclosure relates to the use of fusion proteins containing TGF-beta receptor in combination with taxanes and nucleoside analogues in the preparation of a medicament for the treatment of pancreatic cancer. Specifically, the present disclosure relates to TGF- β receptor-containing fusion proteins comprising an anti-PD-L1 antibody or antigen-binding fragment thereof and a TGF- β receptor moiety, a taxane compound selected from paclitaxel, paclitaxel albumin, docetaxel or a pharmaceutically acceptable salt thereof, a nucleoside analog selected from gemcitabine, mercaptopurine, nelarabine, fludarabine, cladribine, clofarabine, azacitidine, ancitabine, troxacitabine, capecitabine, decitabine.

Description

Application of fusion protein containing TGF-beta receptor in preparation of pancreatic cancer treatment drug by combining with taxoids and nucleoside analogs
Technical Field
The disclosure belongs to the field of pharmacy, and particularly relates to application of a fusion protein containing a TGF-beta receptor in preparation of a medicine for treating pancreatic cancer in combination with a taxoid and a nucleoside analogue.
Background
Programmed death receptor 1 (PD-1) is a member of the CD28 superfamily. PD-1 is expressed in activated T cells, B cells and myeloid cells and has two ligands, programmed death ligand-1 (PD-L1) and PD-L2.PD-L1 interacts with the receptor PD-1 on T cells and plays an important role in the negative regulation of immune responses. The expression of PD-L1 protein can be detected in a plurality of human tumor tissues, the microenvironment of the tumor part can induce the expression of PD-L1 on tumor cells, and the expressed PD-L1 is beneficial to the generation and growth of tumors and induces the apoptosis of anti-tumor T cells. The PD-1/PD-L1 pathway inhibitor can block the combination of PD-1 and PD-L1, block negative regulation signals and restore the activity of T cells, thereby enhancing immune response, so that the immunoregulation taking PD-1/PD-L1 as a target point has important significance for tumor inhibition.
Transforming growth factor-beta (TGF-beta) belongs to the TGF-beta superfamily that regulates cell growth and differentiation. TGF-. Beta.signals through a heterotetrameric receptor complex consisting of two type I and two type II transmembrane serine/threonine kinase receptors.
It has been found that blocking the TGF- β signalling pathway can reduce metastasis of the tumour. The truncated Smad2/3 dominant negative mutant is used for inhibiting TGF-beta signal channel of breast tumor cell line, and the metastatic capability of the tumor cell is found to be inhibited. Microsatellite instability studies of colon cancer have found that inactive mutations in TGF- β RII reduce metastasis and increase patient survival after surgery. However, in general, the effect of the inhibitor for inhibiting TGF-beta signaling pathway used alone in clinical treatment is weak, which may be related to the abnormal high expression of TGF-beta in tumor cells, and the poor efficacy of the inhibitor for inhibiting TGF-beta signaling pathway used alone or the bioavailability of the inhibitor for signaling pathway caused by the difficulty in focusing on tumors.
Therefore, the inhibition of the PD-1/PD-L1 pathway on the basis of targeting and neutralizing TGF-beta of a tumor microenvironment can restore the activity of T cells, enhance immune response and more effectively improve the effect of inhibiting the occurrence and development of tumors. PD-L1/TGF-beta double antibody M-7824 developed by Merck currently enters clinical stage II, and besides being used for treating tumors singly, the product also develops the combination of eribulin mesylate for treating metastatic triple negative breast cancer (NCT 03579472); treatment of previously treated advanced pancreatic adenocarcinoma (NCT 03451773) in combination with gemcitabine; combined with topotecan or temozolomide, the composition is used for treating recurrent small cell lung cancer (NCT 0-3554473) and other indications. WO2018205985A discloses a novel PD-L1/TGF-beta fusion protein.
Pancreatic cancer is highly malignant and progresses rapidly, but has a latent disease and atypical early symptoms, and most patients belong to middle and advanced stages at the time of clinical diagnosis. Treatment of pancreatic cancer is a systematic project, often requiring multidisciplinary consultation Modes (MDT), mainly consisting of surgical, radiation, chemotherapy, interventional and supportive treatment, where surgical resection is the only opportunity for pancreatic cancer to gain a cure and long-term survival. For advanced pancreatic cancer with 1-line treatment failure, the treatment options are limited, and gemcitabine or fluorouracil based single or combination chemotherapy may be selected depending on the patient's physical functional status and tumor progression. PD-1 mAb Pembrolizumab (Pembrolizumab) has currently obtained recommendations of the National Comprehensive Cancer Network (NCCN) guideline class 2 for second line treatment of advanced or metastatic pancreatic cancer patients of the high microsatellite instability (MSI-H)/mismatch repair deficiency (dMMR) type.
Therefore, a more effective treatment new way or a more effective drug combination is searched, and new evidence of evidence-based medicine is provided for the clinical treatment of pancreatic cancer, so that the clinical diagnosis and treatment of patients are guided to be integrated with the current new progress of international and domestic research, and the method has important clinical significance.
Disclosure of Invention
The present disclosure relates to the use of a fusion protein containing TGF-beta receptor in combination with nucleoside analogues and taxoids in the preparation of a medicament for the treatment of tumors or cancers. And a method of treating tumors or cancers by combining a fusion protein containing a TGF-beta receptor with a nucleoside analog, a taxane compound.
In certain embodiments, the nucleoside analog is selected from gemcitabine, mercaptopurine, nelarabine (nelarabine), fludarabine (fludarabine), cladribine (cladribine), clofarabine (clofarabine), azacitidine, ancitabine, troxacitabine, capecitabine, decitabine, or a pharmaceutically acceptable salt of any of the foregoing related drugs. In certain embodiments, the nucleoside analog is gemcitabine, or a pharmaceutically acceptable salt thereof.
In certain embodiments, the taxane compound is selected from paclitaxel, paclitaxel albumin, paclitaxel liposome, docetaxel, or a pharmaceutically acceptable salt thereof. In certain embodiments, paclitaxel albumin.
In certain embodiments, the fusion protein comprising a TGF- β receptor comprises an anti-PD-L1 antibody or antigen-binding fragment thereof and a TGF- β receptor portion, wherein the TGF- β receptor portion is an N-terminal truncated form of the extracellular region of TGF- β RII.
In certain embodiments, the fusion protein comprising a TGF- β receptor is represented by general formula (I):
Ab-L-TGF-βRII ECD (I)
wherein the TGF- β RII ECD is a truncated form of the extracellular domain of TGF- β RII;
ab is an anti-PD-L1 antibody or antigen-binding fragment thereof;
l is a connecting sequence.
In some embodiments, the linking sequence is (G) 4 S) xG, where x is 3 to 6, preferably 3, 4, 5, 6, for example 4.
In certain embodiments, the anti-PD-L1 antibody or antigen-binding fragment thereof comprises:
HCDR1, HCDR2 and HCDR3 shown as SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, respectively, and LCDR1, LCDR2 and LCDR3 shown as SEQ ID NO. 4, SEQ ID NO. 5 and SEQ ID NO. 6, respectively,
wherein, the CDR sequences are shown as follows:
Figure BDA0003863460880000031
wherein X1 is G and X2 is F.
In certain embodiments, the anti-PD-L1 antibody or antigen-binding fragment thereof is a chimeric antibody or a functional fragment thereof, a humanized antibody or a functional fragment thereof, or a human antibody or a functional fragment thereof.
In certain embodiments, the sequences of the heavy and light chains of the humanized anti-PD-L1 antibody are as follows:
anti-PD-L1 antibody heavy chain variable region:
Figure BDA0003863460880000032
anti-PD-L1 antibody light chain variable region:
Figure BDA0003863460880000033
note: the sequence is FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4, and the italic in the sequence is an FR sequence; the CDR sequences are underlined.
In certain embodiments, the heavy chain amino acid sequence of the anti-PD-L1 antibody or antigen-binding fragment thereof is as set forth in SEQ ID NO:9 or a sequence corresponding to SEQ ID NO:9, has at least 85%,86%,87%,88%,89%,90%,91%,92%,93%,94%,95%,96%,97%,98%,99% or 100% sequence identity to the light chain amino acid sequence of the anti-PD-L1 antibody or antigen-binding fragment thereof as set forth in SEQ ID NO:10 or a sequence identical to SEQ ID NO:10 has at least 85%,86%,87%,88%,89%,90%,91%,92%,93%,94%,95%,96%,97%,98%,99% or 100% sequence identity, wherein
anti-PD-L1 antibody heavy chain sequence: igG4 (AA) (S228P)
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYWMHWVRQAPGQGLEWMGRIGPNSGFTSYNEKFKNRVT MTRDTSTSTVYMELSSLRSEDTAVYYCARGGSSYDYFDYWGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEAAGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGA
SEQ ID NO:9
anti-PD-L1 antibody light chain sequence:
DIVLTQSPASLAVSPGQRATITCRASESVSIHGTHLMHWYQQKPGQPPKLLIYAASNLESGVPARFSGS GSGTDFTLTINPVEAEDTANYYCQQSFEDPLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
SEQ ID NO:10
in the present disclosure, the N-terminal truncated form of the TGF- β RII extracellular region is selected from deletions of less than 26 consecutive amino acids, such as from 14 to 26 consecutive amino acids, for example from 14 to 21 consecutive amino acids, for example comprising from 14 to 21 consecutive amino acids, on the N-terminus of the TGF- β RII extracellular region.
In certain embodiments, the sequence of the extracellular region of TGF- β RII is as set forth in SEQ ID NO:11, for example comprising SEQ ID NO: 12. 13 or 14, for example comprising the sequence shown in SEQ ID NO:12, or a sequence shown in figure 12.
Non-limiting example sequences of TGF- β RII extracellular domains and truncated forms thereof in this disclosure are as follows:
TGF- β RII extracellular domain sequence: ECD (1-136)
IPPHVQKSVNNDMIVTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD
SEQ ID NO:11
The TGF- β RII extracellular domain sequence has a 19 amino acid truncation or deletion at the N-terminus: ECD (20-136)
GAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD
SEQ ID NO:12
The TGF-. Beta.RII extracellular domain sequence has a truncation or deletion of 21 amino acids at the N-terminus: ECD (22-136)
VKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD
SEQ ID NO:13
The TGF- β RII extracellular domain sequence has a 14 amino acid truncation or deletion at the N-terminus: ECD (15-136)
VTDNNGAVKFPQLCKFCDVRFSTCDNQKSCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKEKKKPGETFFMCSCSSDECNDNIIFSEEYNTSNPD
SEQ ID NO:14
Passing the heavy chain C-terminal amino acid of the anti-PD-L1 antibody through using homologous recombination technology (G) 4 S) x G is connected with TGF-beta RII extracellular regions with different lengths, and the fusion proteins shown in the table 1 are obtained by carrying out conventional expression by a 293 expression system together with a light chain:
TABLE 1 anti-PD-L1 antibody/TGF-. Beta.RII extracellular region fusion protein
Examples of fusion proteins Description of sequences Number of consecutive amino acid deletions at N-terminus
Fusion protein 1 Ab-(G 4 S) 4 G-ECD(1-136) Is not missing
Fusion protein 2 Ab-(G 4 S) 3 G-ECD(15-136) 14
Fusion protein 3 Ab-(G 4 S) 3 G-ECD(15-136,N19A) 14
Fusion protein 4 Ab-(G 4 S) 3 G-ECD(20-136) 19
Fusion protein 5 Ab-(G 4 S) 3 G-ECD(22-136) 21
Fusion protein 6 Ab-(G 4 S) 3 G-ECD(27-136) 26
Fusion protein 7 Ab-(G4S) 4 G-ECD(15-136) 14
Fusion protein 8 Ab-(G4S) 4 G-ECD(15-136,N19A) 14
Fusion protein 9 Ab-(G 4 S) 4 G-ECD(20-136) 19
Fusion protein 10 Ab-(G 4 S) 4 G-ECD(22-136) 21
Fusion protein 11 Ab-(G 4 S) 4 G-ECD(27-136) 26
Fusion protein 12 Ab-(G 4 S) 5 G-ECD(15-136) 14
Fusion protein 13 Ab-(G 4 S) 5 G-ECD(15-136,N19A) 14
Fusion protein 14 Ab-(G 4 S) 5 G-ECD(20-136) 19
Fusion protein 15 Ab-(G 4 S) 5 G-ECD(22-136) 21
Fusion protein 16 Ab-(G 4 S) 5 G-ECD(27-136) 26
Fusion protein 17 Ab-(G 4 S) 6 G-ECD(27-136) 26
Note: ab is the anti-PD-L1 antibody disclosed by the disclosure, ECD (n-136) in sequence description is the full-length or truncated form of TGF-beta RII extracellular region, and n is the initial number of amino acids after the TGF-beta RII extracellular region is truncated. N19A represents that the 19 th amino acid of the TGF-beta RII extracellular region is mutated into A.
PD-L1/TGF-beta fusion proteins, including their structures and methods of preparation, are herein incorporated in their entirety into WO 2018205985A.
In certain embodiments, the heavy chain amino acid sequence of an anti-PD-L1 antibody or antigen-binding fragment thereof in a fusion protein comprising a TGF-beta receptor is set forth in SEQID NO:9 and the light chain amino acid sequence is shown as SEQ ID NO:10, the linker sequence L is (G) 4 S) 4 G, N-terminal truncated form of TGF-beta RII extracellular region is shown in SEQ ID NO: shown at 12.
In certain embodiments, the anti-PD-L1 antibody or antigen-binding fragment thereof is selected from the group consisting of avelumab, atezolizumab, durvalumab, CS-1001, M-7824, KL-A167, CX-072, BGB-A333, GNS-1480, CA-170, BMS-936559, preferably avelumab, atezolizumab, durvalumab.
In certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered in a dose selected from the group consisting of 1-1600mg/m 2 For example: 10mg/m 2 、15mg/m 2 、20mg/m 2 、25mg/m 2 、30mg/m 2 、35mg/m 2 、40mg/m 2 、45mg/m 2 、50mg/m 2 、55mg/m 2 、60mg/m 2 、65mg/m 2 、70mg/m 2 、75mg/m 2 、80mg/m 2 、85mg/m 2 、90mg/m 2 、95mg/m 2 、100mg/m 2 、105mg/m 2 、110mg/m 2 、115mg/m 2 、120mg/m 2 、125mg/m 2 、130mg/m 2 、135mg/m 2 、140mg/m 2 、145mg/m 2 、150mg/m 2 、155mg/m 2 、160mg/m 2 、165mg/m 2 、170mg/m 2 、175mg/m 2 、180mg/m 2 、185mg/m 2 、190mg/m 2 、195mg/m 2 、200mg/m 2 、210mg/m 2 、220mg/m 2 、230mg/m 2 、240mg/m 2 、250mg/m 2 、260mg/m 2 、270mg/m 2 、280mg/m 2 、290mg/m 2 、300mg/m 2 、310mg/m 2 、320mg/m 2 、330mg/m 2 、340mg/m 2 、350mg/m 2 、360mg/m 2 、370mg/m 2 、380mg/m 2 、390mg/m 2 、400mg/m 2 、410mg/m 2 、420mg/m 2 、430mg/m 2 、440mg/m 2 、450mg/m 2 、460mg/m 2 、470mg/m 2 、480mg/m 2 、490mg/m 2 、500mg/m 2 、510mg/m 2 、520mg/m 2 、530mg/m 2 、540mg/m 2 、550mg/m 2 、560mg/m 2 、570mg/m 2 、580mg/m 2 、590mg/m 2 、600mg/m 2 、625mg/m 2 、650mg/m 2 、675mg/m 2 、700mg/m 2 、725mg/m 2 、750mg/m 2 、775mg/m 2 、800mg/m 2 、825mg/m 2 、850mg/m 2 、875mg/m 2 、900mg/m 2 、925mg/m 2 、950mg/m 2 、975mg/m 2 、1000mg/m 2 、1025mg/m 2 、1050mg/m 2 、1075mg/m 2 、1100mg/m 2 、1125mg/m 2 、1150mg/m 2 、1175mg/m 2 、1200mg/m 2 、1225mg/m 2 、1250mg/m 2 、1275mg/m 2 、1300mg/m 2 、1325mg/m 2 、1350mg/m 2 、1375mg/m 2
1400mg/m 2 、1425mg/m 2 、1450mg/m 2 、1475mg/m 2 、1500mg/m 2 、1525mg/m 2 、1550mg/m 2 、1575mg/m 2 、1600mg/m 2 Or any value in between, preferably 400mg/m 2 、600mg/m 2 、800mg/m 2 、1000mg/m 2 、1200mg/m 2 、1400mg/m 2 、1600mg/m 2 Or any value in between, more preferably 1000mg/m 2 . In certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered twice daily, once every two days, once every three days, once every four days, once every five days, once every six days, once weekly, once every two weeks, once every three weeks, once every four weeks, twice weekly or twice weekly, preferably twice weekly.
In certain embodiments, a nucleoside analog (e.g., gemcitabine is administered at least once during a cycle. In certain embodiments, a nucleoside analog (e.g., gemcitabine) is administered at least twice during a cycle. In certain embodiments, a nucleoside analog (e.g., gemcitabine) is administered at least three times during a cycle. In certain embodiments, a cycle is 14 days. In certain embodiments, a cycle is 21 days. In certain embodiments, a cycle is 28 days. In certain embodiments, gemcitabine is administered at least once weekly. In certain embodiments, in certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered at least once every two weeks, in certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered at least once every three weeks, in certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered at least twice every three weeks, in certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered at least once every four weeks, in certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered on days 1, 8, and 15 of a 28-day cycle, in certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered on days 1, 8 of a 21-day cycle.
In certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered at a dose of 400mg/m 2 、600mg/m 2 、800mg/m 2 、1000mg/m 2 、1200mg/m 2 、1400mg/m 2 Or 1600mg/m 2 The frequency of administration was twice every three weeks. In certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered at a dose of 1000mg/m 2 The frequency of administration was twice every three weeks. In certain embodiments, the nucleoside analog (e.g., gemcitabine) is administered twice every three weeks, on days 1 and 8, respectively.
In certain embodiments, the taxane compound is paclitaxel albumin.
In certain embodiments, the paclitaxel albumin has a weight ratio of albumin to paclitaxel of 1 to 18. In certain embodiments, the paclitaxel albumin has a weight ratio of albumin to paclitaxel of 1. In certain embodiments, the paclitaxel albumin has a weight ratio of albumin to paclitaxel of 1 to 9, for example 9:1. in certain embodiments, the paclitaxel albumin has a weight ratio of albumin to paclitaxel of 1. In certain embodiments, the paclitaxel albumin has a weight ratio of albumin to paclitaxel of 1 to 2.
In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered in a dose selected from the group consisting of 10 to 400mg/m 2 For example: 10mg/m 2 、15mg/m 2 、20mg/m 2 、25mg/m 2 、30mg/m 2 、35mg/m 2 、40mg/m 2 、45mg/m 2 、50mg/m 2 、55mg/m 2 、60mg/m 2 、65mg/m 2 、70mg/m 2 、75mg/m 2 、80mg/m 2 、85mg/m 2 、90mg/m 2 、95mg/m 2 、100mg/m 2 、105mg/m 2 、110mg/m 2 、115mg/m 2 、120mg/m 2 、125mg/m 2 、130mg/m 2 、135mg/m 2 、140mg/m 2 、145mg/m 2 、150mg/m 2 、155mg/m 2 、160mg/m 2 、165mg/m 2 、170mg/m 2 、175mg/m 2 、180mg/m 2 、185mg/m 2 、190mg/m 2 、195mg/m 2 、200mg/m 2 、210mg/m 2 、220mg/m 2 、230mg/m 2 、240mg/m 2 、250mg/m 2 、260mg/m 2 、270mg/m 2 、280mg/m 2 、290mg/m 2 、300mg/m 2 、310mg/m 2 、320mg/m 2 、330mg/m 2 、340mg/m 2 、350mg/m 2 、360mg/m 2 、370mg/m 2 、380mg/m 2 、390mg/m 2 、400mg/m 2 Or any value in between.
In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered in a dose of 25 to 300mg/m 2 For example 50mg/m 2 、75mg/m 2 、100mg/m 2 、125mg/m 2 、150mg/m 2 、175mg/m 2 、200mg/m 2 Or any value in between. In some toolsIn one embodiment, the taxane compound (e.g., paclitaxel albumin) is administered at a dose of 125mg/m 2
In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered twice daily, once every two days, once every three days, once every four days, once every five days, once every six days, once weekly, once every two weeks, once every three weeks, once every four weeks, twice weekly, twice every two weeks, twice every three weeks, or twice every four weeks, preferably twice every three weeks.
In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at a dose of 50mg/m 2 、75mg/m 2 、100mg/m 2 、125mg/m 2 、150mg/m 2 、175mg/m 2 Or 200mg/m 2 Preferably 125mg/m 2 The frequency of administration was twice every three weeks. In certain embodiments, the taxane (e.g., paclitaxel albumin) is administered twice every three weeks, on days 1 and 8, respectively.
In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least once during a cycle. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least twice during a cycle. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least three times during a cycle. In certain embodiments, one cycle is 14 days. In certain embodiments, one cycle is 21 days. In certain embodiments, one cycle is 28 days. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least once per week. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least once every two weeks. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least once every three weeks. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least twice every three weeks. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered at least once every four weeks. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered on days 1, 8, and 15 of a 28 day cycle. In certain embodiments, the taxane compound (e.g., paclitaxel albumin) is administered on days 1, 8 of a 21-day cycle.
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose selected from the group consisting of 0.1-500mg/kg, for example, 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, 8mg/kg, 9mg/kg, 10mg/kg, 11mg/kg, 12mg/kg, 13mg/kg, 14mg/kg, 15mg/kg, 16mg/kg, 17mg/kg, 18mg/kg, 19mg/kg, 20mg/kg, 21mg/kg, 22mg/kg, 23mg/kg, 24mg/kg, 25mg/kg, 26mg/kg, 27mg/kg, 28mg/kg, 29mg/kg, 30mg/kg, 31mg/kg, 32mg/kg, 33mg/kg, 34mg/kg, 35mg/kg, 36mg/kg, 37mg/kg, 38mg/kg, 39mg/kg, 40mg/kg 41mg/kg, 42mg/kg, 43mg/kg, 44mg/kg, 45mg/kg, 46mg/kg, 47mg/kg, 48mg/kg, 49mg/kg, 50mg/kg, 51mg/kg, 52mg/kg, 53mg/kg, 54mg/kg, 55mg/kg, 56mg/kg, 57mg/kg, 58mg/kg, 59mg/kg, 60mg/kg, 61mg/kg, 62mg/kg, 63mg/kg, 64mg/kg, 65mg/kg, 66mg/kg, 67mg/kg, 68mg/kg, 69mg/kg, 70mg/kg, 71mg/kg, 72mg/kg, 73mg/kg, 74mg/kg, 75mg/kg, 76mg/kg, 77mg/kg, 78mg/kg, 79mg/kg, 80mg/kg, 81mg/kg, 82mg/kg, 83mg/kg, 84mg/kg, 85mg/kg, 86mg/kg, 87mg/kg, 88mg/kg, 89mg/kg, 90mg/kg, 91mg/kg, 92mg/kg, 93mg/kg, 94mg/kg, 95mg/kg, 96mg/kg, 97mg/kg, 98mg/kg, 99mg/kg, 100mg/kg, 105mg/kg, 110mg/kg, 115mg/kg, 120mg/kg, 125mg/kg, 130mg/kg, 135mg/kg, 140mg/kg, 145mg/kg, 150mg/kg, 155mg/kg, 160mg/kg, 165mg/kg, 170mg/kg, 175mg/kg, 180mg/kg, 185mg/kg, 190mg/kg, 195mg/kg 200mg/kg, 205mg/kg, 210mg/kg, 215mg/kg, 220mg/kg, 225mg/kg, 230mg/kg, 235mg/kg, 240mg/kg, 245mg/kg, 250mg/kg, 260mg/kg, 270mg/kg, 280mg/kg, 290mg/kg, 300mg/kg, 310mg/kg, 320mg/kg, 330mg/kg, 340mg/kg, 350mg/kg, 360mg/kg, 370mg/kg, 380mg/kg, 390mg/kg, 400mg/kg, 410mg/kg, 420mg/kg, 430mg/kg, 440mg/kg, 450mg/kg, 460mg/kg, 470mg/kg, 480mg/kg, 490mg/kg, 500mg/kg or any value therebetween, preferably 1mg/kg, 3mg/kg, 10mg/kg, 15mg/kg, 20mg/kg, 25mg/kg, 30mg/kg, 35mg/kg, 40mg/kg, 45mg/kg, 50mg/kg, 55mg/kg, 60mg/kg or any value therebetween, more preferably 10mg/kg, 20mg/kg, 30mg/kg, 40mg/kg, 50mg/kg, 60mg/kg or any value therebetween, most preferably 20mg/kg or 30mg/kg. In certain embodiments, the fusion protein comprising a TGF- β receptor is administered weekly, biweekly, every three weeks or every four weeks, preferably every three weeks.
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose selected from 0.1 to 200mg/kg at a frequency of once per week, once every two weeks, once every three weeks, or once every four weeks.
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose selected from the group consisting of 0.1 to 100mg/kg at a frequency of once per week, once every two weeks, once every three weeks, or once every four weeks.
<xnotran> , TGF- β 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, 8mg/kg, 9mg/kg, 10mg/kg, 11mg/kg, 12mg/kg, 13mg/kg, 14mg/kg, 15mg/kg, 16mg/kg, 17mg/kg, 18mg/kg, 19mg/kg, 20mg/kg, 21mg/kg, 22mg/kg, 23mg/kg, 24mg/kg, 25mg/kg, 26mg/kg, 27mg/kg, 28mg/kg, 29mg/kg, 30mg/kg, 31mg/kg, 32mg/kg, 33mg/kg, 34mg/kg, 35mg/kg, 36mg/kg, 37mg/kg, 38mg/kg, 39mg/kg, 40mg/kg, 41mg/kg, 42mg/kg, 43mg/kg, 44mg/kg, 45mg/kg, 46mg/kg, 47mg/kg, 48mg/kg, 49mg/kg, 50mg/kg, 51mg/kg, 52mg/kg, 53mg/kg, 54mg/kg, 55mg/kg, 56mg/kg, 57mg/kg, 58mg/kg, 59mg/kg, 60mg/kg, , . </xnotran>
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose selected from the group consisting of 1mg/kg, 3mg/kg, 10mg/kg, 15mg/kg, 20mg/kg, 25mg/kg, 30mg/kg, 35mg/kg, 40mg/kg, 45mg/kg, 50mg/kg, 55mg/kg, 60mg/kg at a frequency of once every two weeks, once every three weeks, or once every four weeks.
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose selected from the group consisting of 10mg/kg, 20mg/kg, 30mg/kg, 40mg/kg, 50mg/kg, 60mg/kg at a frequency of once every two weeks, once every three weeks, or once every four weeks.
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose of 20mg/kg, 30mg/kg, once every three weeks, on day 1.
<xnotran> , TGF- β 1-4000mg, 25mg, 50mg, 75mg, 100mg, 150mg, 200mg, 250mg, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, 800mg, 850mg, 900mg, 950mg, 1000mg, 1050mg, 1100mg, 1150mg, 1200mg, 1250mg, 1300mg, 1350mg, 1400mg, 1450mg, 1500mg, 1550mg, 1600mg, 1650mg, 1700mg, 1750mg, 1800mg, 1850mg, 1900mg, 1950mg, 2000mg, 2050mg, 2100mg, 2150mg, 2200mg, 2250mg, 2300mg, 2350mg, 2400mg, 2450mg, 2500mg, 2550mg, 2600mg, 2650mg, 2700mg, 2750mg, 2800mg, 2850mg, 2900mg, 2950mg, 3000mg, 3050mg, 3100mg, 3150mg, 3200mg, 3250mg, 3300mg, 3350mg, 3400mg, 3450mg, 3500mg, 3550mg, 3600mg, 3650mg, 3700mg, 3750mg, 3800mg, 3850mg, 3900mg, 3950mg, 4000mg , 300mg, 600mg, 900mg, 1200mg, 1500mg, 1800mg, 2100mg, 2400mg, 2700mg, 3000mg, 3300mg, 3600mg , 600mg, 1200mg, 1800mg, 2400mg, 3000mg . </xnotran> In certain embodiments, the frequency of administration is once a week, once every two weeks, once every three weeks, or once every four weeks, preferably once every two weeks or once every three weeks, more preferably once every three weeks.
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose of 300mg, 600mg, 900mg, 1200mg, 1500mg, 1800mg, 2100mg, 2400mg, 2700mg, 3000mg, 3300mg, 3600mg, at a frequency of once every two weeks, once every three weeks, or once every four weeks.
In certain embodiments, the fusion protein comprising a TGF- β receptor is administered at a dose of 600mg, 1200mg, 1800mg, 2400mg, 3000mg, at a frequency of once every two weeks or once every three weeks. In certain embodiments, the fusion protein comprising a TGF- β receptor is administered once every three weeks on day 1.
In alternative embodiments, the fusion protein containing a TGF- β receptor is administered by injection, such as subcutaneous or intravenous injection, the fusion protein containing a TGF- β receptor is formulated into an injectable form prior to injection, and the injectable form of the fusion protein containing a TGF- β receptor may be an injectable solution or a lyophilized powder, which comprises the fusion protein containing a TGF- β receptor, a buffer, a stabilizer, and optionally a surfactant. The buffer can be one or more selected from acetate, citrate, succinate and phosphate. The stabilizer may be selected from sugars or amino acids, preferably disaccharides, such as sucrose, lactose, trehalose, maltose. The surfactant is selected from polyoxyethylene hydrogenated castor oil, glycerin fatty acid ester, polyoxyethylene sorbitan fatty acid ester, etc.
The present disclosure provides a method of treating a tumor or cancer comprising administering to a patient an effective amount of the above nucleoside analog, a taxane compound, and the above effective amount of a fusion protein comprising a TGF- β receptor.
In alternative embodiments, the patient is a human, provided herein is a method of treating a tumor or cancer.
The tumor or cancer in the present disclosure is selected from the group consisting of tumors or cancers of: colorectal, breast, ovarian, pancreatic, gastric, prostate, renal, cervical, myeloma, lymphoma, leukemia, thyroid, endometrial, uterine, bladder, neuroendocrine, head and neck, liver, nasopharyngeal, testicular, small cell lung cancer, non-small cell lung cancer, melanoma, basal cell skin cancer, squamous cell skin cancer, dermatofibrosarcoma protruberans, merkel cell cancer, glioblastoma, glioma, sarcoma, mesothelioma, and myelodysplastic syndrome.
In certain embodiments, the tumor or cancer is pancreatic cancer.
In certain embodiments, the pancreatic cancer is advanced pancreatic cancer.
In certain embodiments, the pancreatic cancer is metastatic pancreatic cancer.
In certain embodiments, the pancreatic cancer is ductal adenocarcinoma or acinar cell carcinoma.
In alternative embodiments of the disclosure, the pancreatic cancer is not chemotherapy. In certain other embodiments, the pancreatic cancer is chemotherapy.
In an alternative embodiment of the present disclosure, wherein the chemotherapeutic agent is selected from at least one of platinum group drugs, epirubicin, 5-FU, irinotecan, nucleoside analogs or taxanes.
Platinum drugs described in the present disclosure include, but are not limited to, carboplatin, cisplatin, oxaliplatin, nedaplatin (Nedaplatin), lobaplatin (lobaplatin), satraplatin (satraplatin), cycloplatin (cycloplatin), miboplatin (Miboplatin), enloplatin, iproplatin, and Dicycloplatin.
As used herein, a "combination" or "co-administration" of a drug means that at least one dose of a taxane compound, a nucleoside analog, and at least one dose of a TGF- β receptor containing fusion protein are administered over a period of time, wherein both substances exhibit pharmacological effects. The time period may be within one administration cycle, optionally within 4 weeks, within 3 weeks, within 2 weeks, within 1 week, within 24 hours, within 2 hours. The taxanes, nucleoside analogs and fusion proteins containing TGF-beta receptors may be administered simultaneously or sequentially. Such terms include treatments in which the taxane, nucleoside analog, and fusion protein containing the TGF- β receptor are administered by the same route of administration or by different routes of administration. The modes of administration of the combinations of the present disclosure are selected from simultaneous administration, independently formulated and co-administered, or independently formulated and administered sequentially.
In certain embodiments, one treatment cycle every two weeks, one treatment cycle every three weeks or one treatment cycle every four weeks, preferably one treatment cycle every three weeks.
In certain embodiments, the drug treatment is administered on day 1, day 8, and/or day 15 of each cycle.
In certain embodiments, the drug treatment is administered on days 1, 8 of each cycle.
In certain embodiments, the fusion protein containing the TGF- β receptor, the taxane and the nucleoside analog are administered on day 1 of each cycle, and only the taxane and nucleoside analog treatment is administered on day 8; in certain more specific embodiments, the fusion protein comprising a TGF- β receptor, the taxane and the nucleoside analog are administered simultaneously or sequentially.
In certain embodiments, an intravenous drip of the fusion protein containing the TGF- β receptor is administered, followed by administration of the taxane, and finally administration of the nucleoside analog. In certain embodiments, an intravenous drip of the fusion protein containing the TGF- β receptor is administered, followed by administration of the nucleoside analog, and finally administration of the taxane compound. In certain embodiments, an intravenous drip of the fusion protein containing the TGF- β receptor is administered prior to the simultaneous administration of the nucleoside analog and the taxane.
In certain embodiments, the nucleoside analog is administered by intravenous drip, followed by administration of the taxane compound, and finally administration of the fusion protein comprising the TGF- β receptor. In certain embodiments, the nucleoside analog is administered by intravenous drip, followed by administration of the fusion protein comprising the TGF- β receptor, and finally administration of the taxane compound. In certain embodiments, the nucleoside analog is administered by intravenous drip prior to the concurrent administration of the fusion protein comprising the TGF- β receptor and the taxane.
In certain embodiments, the taxane compound is administered by intravenous drip, followed by administration of the nucleoside analog, and finally administration of the fusion protein comprising the TGF- β receptor. In certain embodiments, the taxane compound is administered by intravenous drip, followed by administration of the fusion protein comprising the TGF- β receptor, and finally the nucleoside analog. In certain embodiments, the taxane compound is administered by intravenous drip prior to the simultaneous administration of the nucleoside analog and the fusion protein comprising the TGF- β receptor.
In certain embodiments, each drug may be administered at least 30min apart.
In certain specific embodiments, each three weeks is a treatment cycle, with sequential administration of drug therapy on day 1 of each cycle, by intravenous drip of TGF- β receptor fusion protein, at least 30min apart, followed by paclitaxel albumin and finally gemcitabine; paclitaxel albumin was administered prior to gemcitabine on day 8 of each cycle.
In certain embodiments, the TGF- β receptor fusion protein and the chemotherapeutic agent are as consistent as possible.
The term "antibody" as used herein refers to an immunoglobulin, which is a tetrapeptide chain structure formed by two identical heavy chains and two identical light chains linked by interchain disulfide bonds.
In the present disclosure, the antibody light chain of the present disclosure may further comprise a light chain constant region comprising a human or murine kappa, lambda chain or variant thereof.
In the present disclosure, the antibody heavy chain of the present disclosure may further comprise a heavy chain constant region comprising IgG1, igG2, igG3, igG4 of human or murine origin, or a variant thereof.
The approximately 110 amino acid sequence of the heavy and light chains of antibodies near the N-terminus varies widely, as the variable region (Fv region); the remaining amino acid sequence near the C-terminus is relatively stable and is a constant region. The variable regions include 3 hypervariable regions (HVRs) and 4 Framework Regions (FRs) with relatively conserved sequences. The 3 hypervariable regions determine the specificity of the antibody, also known as Complementarity Determining Regions (CDRs). Each Light Chain Variable Region (LCVR) and Heavy Chain Variable Region (HCVR) is composed of 3 CDR regions and 4 FR regions, arranged sequentially from amino terminus to carboxy terminus in the order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The 3 CDR regions of the light chain refer to LCDR1, LCDR2, and LCDR3; the 3 CDR regions of the heavy chain refer to HCDR1, HCDR2 and HCDR3. The CDR amino acid residues of the LCVR and HCVR regions of the antibodies or antigen-binding fragments of the present disclosure are in number and position in accordance with known Kabat numbering convention (LCDR 1-3, HCDR1-3), or in accordance with Kabat and chothia numbering convention (HCDR 1).
Antibodies of the present disclosure include murine, chimeric, humanized, preferably humanized antibodies.
The term "antibody or antigen-binding" or "functional fragment" thereof as used in this disclosure refers to Fab fragments, fab 'fragments, F (ab') 2 fragments, and Fv fragment ScFv fragments that bind to antibodies, which have antigen-binding activity. The Fv fragment contains the variable regions of the antibody heavy and light chains, but no constant regions, and has the smallest antibody fragment of the entire antigen-binding site. Generally, fv antibodies also comprise a polypeptide linker between the VH and VL domains and are capable of forming the structures required for antigen binding. Two antibody variable regions can also be joined together with different linkers into a single polypeptide chain, known as single chain antibodies (scFv) or single chain Fv (sFv). The term "binds to PD-L1" in the present disclosure means capable of interacting with human PD-L1. The term "antigen binding site" of the present disclosure refers to a three-dimensional spatial site that is not contiguous on an antigen and is recognized by an antibody or antigen binding fragment of the present disclosure.
The term "murine antibody" is used in this disclosure to refer to a monoclonal antibody to human PD-L1 prepared according to the knowledge and skill in the art. Preparation is performed by injecting the test subject with the PD-L1 antigen and then isolating hybridomas that express antibodies having the desired sequence or functional properties.
The term "chimeric antibody" refers to an antibody obtained by fusing a variable region of a murine antibody to a constant region of a human antibody, and can reduce an immune response induced by the murine antibody. Establishing chimeric antibody, firstly establishing hybridoma secreting mouse-derived specific monoclonal antibody, then cloning variable region gene from mouse hybridoma cell, cloning constant region gene of human antibody according to the need, connecting mouse variable region gene and human constant region gene into chimeric gene, inserting into human carrier, and finally expressing chimeric antibody molecule in eukaryotic industrial system or prokaryotic industrial system. In a preferred embodiment of the present disclosure, the antibody light chain of the PCSK-9 chimeric antibody further comprises a light chain constant region of a human kappa, lambda chain or variant thereof. The antibody heavy chain of the PCSK-9 chimeric antibody further comprises a heavy chain constant region of a human IgG1, igG2, igG3, igG4 or a variant thereof. The constant region of the human antibody may be selected from the heavy chain constant region of human IgG1, igG2, igG3 or IgG4 or a variant thereof, preferably comprising human IgG2 or IgG4 heavy chain constant region, or IgG4 that is free of ADCC (antibody-dependent cell-mediated cytotoxicity) toxicity after amino acid mutation using an amino acid.
The term "humanized antibody", also known as CDR-grafted antibody (CDR), refers to an antibody produced by grafting mouse CDR sequences into a human antibody variable region framework, i.e., a different type of human germline antibody framework sequence. Can overcome the strong antibody variable antibody reaction induced by the chimeric antibody because of carrying a large amount of mouse protein components. Such framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences. Germline DNA sequences of, for example, human heavy and light chain variable region genes can be found in the "VBase" human germline sequence database (at the Internet)www.mrccpe.com.ac.uk/vbaseAvailable), and found in Kabat, e.a. et al, 1991Sequences of Proteins of Immunological Interest, 5 th edition. To avoid reduced immunogenicity and reduced activity, the human antibody variable region framework sequences may be minimally back-mutated or back-mutated to retain activity. Humanized antibodies of the present disclosure also include humanized antibodies after further affinity maturation of the CDRs by phage display.
The term "identity" in this disclosure refers to sequence similarity between two polynucleotide sequences or between two polypeptides. Sequence identity in the present disclosure may be at least 85%, 90% or 95%, preferably at least 95%. Non-limiting examples include 85%,86%,87%,88%,89%,90%,91%,92%,93%,94%,95%,96%,97%,98%,99%,100%. Sequence comparison and percent identity determination between two sequences can be performed by the default settings of the BLASTN/BLASTP algorithm available on the National Center For Biotechnology Institute website.
The term "TGF- β receptor II" or "TGF β RII" or "transforming growth factor β receptor II" refers to a cell surface receptor that binds to a ligand (including but not limited to TGF β 1, TGF β 2, and TGF β 3) and thereby initiates a signal transduction pathway within a cell.
The term "PD-L1" refers to programmed death ligand 1, also known as CD274 and B7H1.PD-L1 is a 290 amino acid protein with an extracellular IgV-like and IgC-like domain (amino acids 19-239 of full-length PD-L1), a transmembrane domain and an intracellular domain of about 30 amino acids. PD-L1 is constitutively expressed on many cells such as antigen presenting cells (e.g., dendritic cells, macrophages, and B cells) as well as hematopoietic and non-hematopoietic cells (e.g., vascular endothelial cells, pancreatic islets, and sites of immune privilege). PD-L1 is also expressed on a variety of tumor and virus infected cells and is a component of the immunosuppressive environment (immunosuppressive milieu) (Ribas 2012, nejm 366. PD-L1 binds to one of two T cell co-inhibitors PD-1 and B7-1.
The fusion protein described in the present disclosure is a protein product co-expressed by two genes obtained by DNA recombination. Methods for producing and purifying antibodies and antigen-binding fragments are well known in the art, such as the Cold spring harbor antibody protocols, chapters 5-8 and 15. For example, mice can be immunized with human PD-L1 or fragments thereof, and the resulting antibodies can be renatured, purified, and amino acid sequenced using conventional methods. Antigen-binding fragments can likewise be prepared by conventional methods. The antibody or antigen binding fragment of the invention is genetically engineered to add one or more human FR regions to the CDR regions of non-human origin. Human FR germline sequences can be obtained from ImmunoGeneTiCs (IMGT) website http:// IMGT. Cities. FR or from the immunoglobulin journal, 2001ISBN012441351 by aligning the IMGT human antibody variable region germline gene database with the MOE software.
An "effective amount" or "therapeutically effective amount" comprises an amount sufficient to ameliorate or prevent a symptom or condition of a medical condition. An effective amount also means an amount sufficient to allow or facilitate diagnosis. The effective amount for a particular patient or veterinary subject may vary depending on the following factors: for example, the condition to be treated, the overall health of the patient, the method and dosage of administration, and the severity of side effects. An effective amount may be the maximum dose or dosage regimen that avoids significant side effects or toxic effects.
Detailed Description
The present invention is further described below with reference to examples, but these examples do not limit the scope of the present disclosure.
Example 1 evaluation of test drugs for safety and efficacy in humans.
54-60 subjects who have not been treated systemically for advanced/metastatic pancreatic cancer are recruited, and the effect of TGF-beta receptor fusion protein in combination with taxanes and nucleoside analogues in advanced/metastatic pancreatic cancer is initially evaluated.
1. Test antibodies and compounds
Recombinant TGF-beta receptor fusion protein for injection (fusion protein 9)
(manufacturer: suzhou Shengdiya biological medicine Co., ltd., specification: 0.3g/6mL, light-proof, 2-8 ℃ storage and transportation, no freezing or shaking, preparation method refer to the drug handbook): 30mg/kg;20mg/kg
Chemotherapy drugs:
the taxoid compound: paclitaxel Albumin (paclitaxel for injection (albumin-bound type), manufacturer: hengrui pharmaceutical Co., ltd, jiangsu, specification: 100 mg/bottle, light-shielding, storage and transportation at 20-30 deg.C)
Nucleoside analogs: gemcitabine (manufacturer: jiangsu Haofen pharmaceutical Co., ltd., specification: 1.0g, lyophilized powder for injection, sealing, storage in dry place)
2. Criteria for subjects in group
1) Histologically or cytologically confirmed advanced/metastatic pancreatic cancer;
2) Has not been treated systemically for advanced/metastatic pancreatic cancer;
3) Age: is 18-70 years old, and can be used by both men and women.
3. Method of administration
TGF-beta receptor fusion protein: intravenous administration at a dose of 30mg/kg or 20mg/kg, 1 time for 3 weeks, one cycle every 3 weeks, on day 1 of each cycle. It is recommended to use the infusion pump for administration, the administration is completed by intravenous drip for 30-60 minutes, intravenous bolus injection and rapid bolus injection are not needed, and after the infusion is finished, the infusion tube is flushed by sufficient physiological saline or 5% glucose solution.
Gemcitabine: 1000mg/m 2 (initial dose) one dosing cycle every 3 weeks (21 days), with intravenous drip given on days 1, 8 of the cycle.
Paclitaxel albumin: 125mg/m 2 (initial dose) 1 dosing cycle every 3 weeks, given as an intravenous drip on days 1 and 8 of each cycle.
Every three weeks is a treatment cycle, with sequential drug treatments on day 1 of each cycle, i.e., intravenous drip of TGF-beta receptor fusion protein (fusion protein 9), followed by paclitaxel albumin and finally gemcitabine at intervals of at least 30 min. TGF-beta receptor fusion proteins and chemotherapeutic agents remain as consistent as possible.
After the subjects developed a progression that met the criteria for the evaluation of the efficacy of solid tumors as defined in version 1.1 (RECIST 1.1), TGF-beta receptor fusion protein and taxanes, nucleoside analogs therapy could continue if the investigator assessed that the subjects had clinical benefit and could tolerate the study therapy and the patients were voluntary.
Example 2 therapeutic Effect of TGF-beta receptor fusion proteins in combination with taxanes, nucleoside analogs in advanced/metastatic pancreatic cancer
The purpose is as follows: patients with histologically or cytologically confirmed unresectable locally advanced or metastatic pancreatic cancer and never received systemic anti-tumor therapy for advanced or metastatic disease were selected.
The following administration modes are adopted:
TGF-beta receptor fusion protein 9: the dose was 30mg/kg, 1 administration cycle every 3 weeks, and 1 day of administration per cycle was given by intravenous drip.
Gemcitabine: 1000mg/m 2 Every 3 weeks for 1 administration cycle, intravenous drip was given on days 1 and 8 of each cycle.
Paclitaxel albumin: 125mg/m 2 Every 3 weeks for 1 administration cycle, intravenous drip was given on days 1 and 8 of each cycle.
The research results are as follows:
no dose-limiting toxicity was observed in the first 6 patients. The RP2D of TGF-beta receptor fusion protein 9 was 30mg/kg.
By the date of data expiration at the time of analysis, a total of 56 patients were enrolled with a median follow-up period of 4.2 months (range: 0.2-10.6 months), of which 28 patients were still receiving treatment. Of the 52 patients who received the imaging assessment at least once after baseline, 19 patients (36.5%, 95% ci 23.6-51.0 confirmed objective remission) achieved objective remission (ORR), 42 patients (80.8%, 95% ci 67.5-90.4) achieved stable Disease Control (DCR). Among the 56 patients receiving at least one dose of study drug treatment, the median progression-free survival (PFS) was 5.5 months (95% CI 4.7-6.9), with a total survival (OS) at 9 months of 62.8% (95% CI 33.6-82.0).
The above results show that TGF-beta receptor fusion protein 9, in combination with gemcitabine and paclitaxel albumin, exhibits anti-tumor activity in untreated locally advanced or metastatic pancreatic cancer. This combination therapy is well tolerated and helps to overcome the failure of at least one dose of the study drug in advanced/metastatic pancreatic cancer.

Claims (23)

1. The use of a fusion protein containing TGF-beta receptor in combination with a taxoid and a nucleoside analogue in the preparation of a medicament for treating pancreatic cancer;
preferably, the pancreatic cancer is advanced or metastatic pancreatic cancer;
preferably, the pancreatic cancer is ductal adenocarcinoma or acinar cell carcinoma.
2. The use according to claim 1, wherein the fusion protein comprising a TGF- β receptor comprises an anti-PD-L1 antibody or antigen-binding fragment thereof and a TGF- β receptor moiety, wherein the TGF- β receptor moiety is an N-terminal truncated form of the extracellular region of TGF- β RII.
3. The use according to claim 2, wherein the fusion protein comprising a TGF- β receptor is of the general formula (I):
Ab-L-TGF-βRII ECD(I)
wherein the TGF-beta RII ECD is a truncated form of the extracellular domain of TGF-beta RII;
ab is an anti-PD-L1 antibody or antigen-binding fragment thereof;
l is a connecting sequence.
4. The use according to claim 3, wherein the linker sequence is (G) 4 S) xG, where x is 3 to 6, preferably 4.
5. The use according to claim 3, wherein the sequence of the extracellular domain of TGF- β RII is as set forth in SEQ ID NO:11 is shown in the figure; preferably comprises SEQ ID NO: 12. 13 and 14, more preferably a sequence comprising SEQ ID NO:12, or a sequence shown in figure 12.
6. The use of any one of claims 2-5, the anti-PD-L1 antibody or antigen-binding fragment thereof comprising:
HCDR1, HCDR2 and HCDR3 shown as SEQ ID NO. 1, SEQ ID NO. 2 and SEQ ID NO. 3, respectively, and LCDR1, LCDR2 and LCDR3 shown as SEQ ID NO. 4, SEQ ID NO. 5 and SEQ ID NO. 6, respectively.
7. The use according to any one of claims 2 to 6, wherein the anti-PD-L1 antibody is a chimeric, humanized or human antibody.
8. The use of claim 7, wherein the anti-PD-L1 antibody or antigen-binding fragment thereof comprises a heavy chain variable region as set forth in SEQ ID NO. 7 and a light chain variable region as set forth in SEQ ID NO. 8.
9. The use of any one of claims 2-8, wherein the heavy chain amino acid sequence of the anti-PD-L1 antibody or antigen-binding fragment thereof is as set forth in SEQ ID NO:9 or has at least 90% sequence identity thereto, and the light chain amino acid sequence of the anti-PD-L1 antibody or antigen-binding fragment thereof is as set forth in SEQ ID NO:10 or has at least 90% sequence identity thereto.
10. Use according to any one of claims 2 to 9, of an anti-PD-L1 antibody or of an anti-PD-L1 antibody in a fusion protein comprising a TGF- β receptorThe heavy chain amino acid sequence of the antigen binding fragment is shown as SEQ ID NO:9, and the light chain amino acid sequence is shown as SEQ ID NO:10, the linker sequence L is (G) 4 S) 4 G, N-terminal truncated form of TGF-beta RII extracellular region is shown in SEQ ID NO: shown at 12.
11. Use according to any of claims 1 to 10, wherein the fusion protein comprising a TGF- β receptor is administered at a dose selected from the group consisting of 0.1-500mg/kg, preferably 3mg/kg, 10mg/kg, 15mg/kg, 20mg/kg, 25mg/kg, 30mg/kg, 35mg/kg, 40mg/kg, 45mg/kg, 50mg/kg, 55mg/kg, 60mg/kg, 65mg/kg, 70mg/kg, 75mg/kg, 80mg/kg, 90mg/kg, 100mg/kg, 125mg/kg, 150mg/kg, more preferably 20mg/kg or 30mg/kg.
12. Use according to any one of claims 1 to 11, whereby the fusion protein comprising a TGF- β receptor is administered weekly, biweekly, every three weeks or every four weeks, preferably every three weeks.
13. Use according to any of claims 1 to 12, wherein the fusion protein comprising a TGF- β receptor is administered in a dose selected from the group consisting of 1-4000mg, preferably 100mg, 200mg, 300mg, 600mg, 900mg, 1200mg, 1500mg, 1800mg, 2100mg, 2400mg, 2700mg, 3000mg, 3300mg, 3600mg.
14. Use according to any one of claims 1 to 13, whereby the fusion protein comprising a TGF- β receptor is administered weekly, biweekly, every three weeks or every four weeks, preferably every three weeks.
15. The use according to any one of claims 1 to 14, wherein the nucleoside analogue is selected from the group consisting of: gemcitabine, mercaptopurine, nelarabine (nelarabine), fludarabine (fludarabine), cladribine (cladribine), clofarabine (clofarabine), azacitidine, ancitabine, troxacitabine, capecitabine, decitabine, preferably gemcitabine or a pharmaceutically acceptable salt thereof.
16. The use according to any one of claims 1 to 15, wherein said nucleoside analogue is administered in a dose selected from the group consisting of 1 to 1600mg/m 2 The frequency of administration is twice weekly, twice every three weeks or twice every four weeks;
preferably, the nucleoside analogue is administered at a dose of 400mg/m 2 、600mg/m 2 、800mg/m 2 、1000mg/m 2 、1200mg/m 2 、1400mg/m 2 、1600mg/m 2 Twice every three weeks;
more preferably, the nucleoside analogue is administered at a dose of 1000mg/m 2 Twice every three weeks.
17. The use according to any one of claims 1 to 16, wherein the taxane compound is selected from paclitaxel, paclitaxel albumin, docetaxel or a pharmaceutically acceptable salt thereof, preferably paclitaxel albumin.
18. The use according to any one of claims 1 to 17, wherein the taxoid is administered in a dose selected from the range of 10 to 400mg/m 2 The frequency of administration is twice weekly, twice weekly or twice weekly,
preferably, the taxoid is administered in a dose selected from 50mg/m 2 、75mg/m 2 、100mg/m 2 、125mg/m 2 、150mg/m 2 、175mg/m 2 、200mg/m 2 The dosing frequency is twice every three weeks;
more preferably, the taxoid is administered at a dose of 125mg/m 2 The dosing frequency was twice every three weeks.
19. The use of any one of claims 1-18, wherein the pancreatic cancer is chemotherapy-treated.
20. The use of any one of claims 1-18, wherein the pancreatic cancer is not chemotherapy-naive.
21. The use according to any one of claims 1 to 20, wherein the fusion protein comprising a TGF- β receptor is administered simultaneously or sequentially with the taxane compound, gemcitabine.
22. A pharmaceutical pack for the treatment of pancreatic cancer comprising a TGF- β receptor-containing fusion protein, taxoid and/or nucleoside analogue of any one of claims 1 to 18;
preferably, the pancreatic cancer is advanced or metastatic pancreatic cancer;
preferably, the pancreatic cancer is ductal adenocarcinoma or acinar cell carcinoma.
23. A pharmaceutical composition for the treatment of pancreatic cancer comprising a TGF- β receptor-containing fusion protein, taxoid and/or nucleoside analogue of any one of claims 1 to 18, together with one or more pharmaceutically acceptable excipients, diluents or carriers;
preferably, the pancreatic cancer is advanced or metastatic pancreatic cancer;
preferably, the pancreatic cancer is ductal adenocarcinoma or acinar cell carcinoma.
CN202211173875.2A 2021-09-26 2022-09-26 Application of fusion protein containing TGF-beta receptor and taxus and nucleoside analogue in preparing medicine for treating pancreatic cancer Pending CN115944741A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202111129650 2021-09-26
CN2021111296502 2021-09-26
CN2022101019777 2022-01-27
CN202210101977 2022-01-27

Publications (1)

Publication Number Publication Date
CN115944741A true CN115944741A (en) 2023-04-11

Family

ID=87288353

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211173875.2A Pending CN115944741A (en) 2021-09-26 2022-09-26 Application of fusion protein containing TGF-beta receptor and taxus and nucleoside analogue in preparing medicine for treating pancreatic cancer

Country Status (1)

Country Link
CN (1) CN115944741A (en)

Similar Documents

Publication Publication Date Title
EP3334431B9 (en) 5-bromo-2,6-di-(1h-pyrazol-l-yl)pyrimidin-4-amine for use in the treatment of cancer
US11597768B2 (en) Immunotherapy for hepatocellular carcinoma
JP7436477B2 (en) TGF-β receptor fusion protein pharmaceutical composition and use thereof
JP2023065391A (en) Anti-ror1 antibodies and methods of making and using thereof
CN109963592B (en) Use of PD-1 antibodies in combination with VEGF ligands or VEGF receptor inhibitors for the preparation of a medicament for the treatment of tumors
TW201249460A (en) Use of anti-CD19 maytansinoid immunoconjugate antibody for the treatment of B-cell malignancies symptoms
EP3685855B1 (en) Human anti-vegfr-2/kdr antibodies
JP2015518827A5 (en)
JP2020520912A (en) Treatment of cancer with anti-GITR agonist antibody
WO2021063331A1 (en) USE OF COMBINATION OF EZH2 INHIBITOR AND TGF-β RECEPTOR-CONTAINING FUSION PROTEIN IN PREPARATION OF DRUGS FOR TREATING TUMORS
CN115944741A (en) Application of fusion protein containing TGF-beta receptor and taxus and nucleoside analogue in preparing medicine for treating pancreatic cancer
CN116745322A (en) Combination therapy using anti-fucosyl-GM 1 antibodies
TWI814752B (en) Uses of immunotherapy agents, nucleoside antimetabolites combined with platinum in the preparation of drugs for treating tumor
TW202214287A (en) Use of combination of tgf-β receptor fusion protein and multi-target tyrosine kinase inhibitor in preparation of medicine of tumor treatment
CN112543637B (en) Use of irinotecan in combination with an immune checkpoint inhibitor and 5-FU for the preparation of a medicament for the treatment of a neoplastic disease
CN115245565A (en) Application of fusion protein containing TGF-beta receptor in preparation of medicine for treating non-small cell lung cancer
EP3280732B1 (en) Antibodies binding to canine vascular endothelial growth factor and uses thereof in treating canine angiogenesis-related diseases
KR20200105825A (en) Use of the combination therapy of PD-1 antibody and Afatinib for the treatment of triple negative breast cancer
US20220259316A1 (en) Multiple-Variable Dosage Regimen for the Treatment of Cancers with High Expression of EGFR
CN117224689B (en) Use of a combination of an anti-HER 2 antibody and a chemotherapeutic agent for the treatment of gastric cancer
WO2023051674A1 (en) Anti-cd47 antibody for combined treatment of blood tumor
CA3227172A1 (en) Methods for treating acute myeloid leukemia with anti-ilt3 antibodies
TW201642904A (en) Composition comprising anti-FGFR2 antibody and other agent
WO2016057726A2 (en) Human anti-vegfr-2/kdr antibodies
WO2023183231A1 (en) Combination therapy methods with t-cell engaging molecules for treatment of prostate cancer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication