CN112795647A - Tumor marker and application thereof - Google Patents

Tumor marker and application thereof Download PDF

Info

Publication number
CN112795647A
CN112795647A CN201911117303.0A CN201911117303A CN112795647A CN 112795647 A CN112795647 A CN 112795647A CN 201911117303 A CN201911117303 A CN 201911117303A CN 112795647 A CN112795647 A CN 112795647A
Authority
CN
China
Prior art keywords
c10orf71
mutation
tumor
cancer
related molecule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN201911117303.0A
Other languages
Chinese (zh)
Other versions
CN112795647B (en
Inventor
季加孚
李子禹
高翔宇
季序我
梁晗
彭鑫鑫
李哲
魏斌
韦宝耶
董宇
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.)
Precision Scientific Technology Beijing Co ltd
Beijing Cancer Hospital
Original Assignee
Precision Scientific Technology Beijing Co ltd
Beijing Cancer Hospital
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 Precision Scientific Technology Beijing Co ltd, Beijing Cancer Hospital filed Critical Precision Scientific Technology Beijing Co ltd
Priority to CN201911117303.0A priority Critical patent/CN112795647B/en
Publication of CN112795647A publication Critical patent/CN112795647A/en
Application granted granted Critical
Publication of CN112795647B publication Critical patent/CN112795647B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Pathology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Physics & Mathematics (AREA)
  • Oncology (AREA)
  • Wood Science & Technology (AREA)
  • Urology & Nephrology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • General Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Food Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The invention relates to a tumor marker and application thereof, in particular to application of at least one of a C10orf71 gene, a C10orf71 protein and a C10orf71 gene detection primer kit in preparing a product for predicting response of a cancer patient to neoadjuvant chemotherapy. The invention firstly determines the key molecular characteristics of C10orf71 gene mutation and the response of tumors to neoadjuvant chemotherapy, and based on the technical scheme of the invention, the neoadjuvant chemotherapy has the advantages of improving prognosis and determining responsive patients before the treatment starts, so that a better treatment scheme can be established.

Description

Tumor marker and application thereof
Technical Field
The invention relates to the field of tumor molecular biology, in particular to a tumor cancer marker and application thereof.
Background
Neoadjuvant chemotherapy is a widely used treatment option for cancer patients, and several large clinical trials have shown that neoadjuvant chemotherapy has advantages in improving prognosis and identifying responsive patients before treatment begins, and thus better treatment regimens can be developed. However, a significant proportion of cancer patients respond poorly to neoadjuvant chemotherapy. To date, only certain clinical features (e.g., age and preoperative weight loss) are available for patient selection, but function is very limited. Furthermore, it is unclear how neoadjuvant chemotherapy alters cancer signaling and affects the tumor microenvironment.
Gastric Cancer (GC) is one of the most common types of cancer in the world and is also the second most common cause of cancer-related death (8.2%). China is the most GC-afflicted country, and based on data from the central cancer registry of the chinese country, over 679,000 newly diagnosed GC cases occur, with about 498,000 GC-related deaths occurring each year. Most of these patients are in the advanced stage, with 5-year survival < 30%. Although surgical treatment is the primary treatment for GC, multimodal strategies are employed to improve patient survival. Neoadjuvant (or perioperative) chemotherapy can "shrink the size of the tumor" before attempting radical resection. In addition, neoadjuvant chemotherapy helps reduce risk by eliminating potential cancer cells and providing a sensitive treatment regimen for postoperative adjuvant chemotherapy in GC patients with high risk of distant metastasis. The benefits of this approach compared to surgery alone have been demonstrated in a number of clinical trials. Currently, neoadjuvant chemotherapy is routinely used to eradicate patients with advanced GC.
However, there is still a significant fraction of GC cancer patients who respond poorly to neoadjuvant chemotherapy, and it is unclear how neoadjuvant chemotherapy alters cancer signaling and affects the tumor microenvironment. A thorough understanding of these problems is crucial to developing an optimized multi-modal treatment plan. Therefore, there is an urgent need to characterize molecular markers that can predict therapeutic response.
Disclosure of Invention
In an effort to explore how neoadjuvant chemotherapy alters cancer signaling and affects the tumor microenvironment, the team of inventors succeeded in obtaining molecular markers that can predict therapeutic response. This finding is crucial for the formulation of an optimized multi-modal treatment plan for cancer patients. In accordance with this, the first and second electrodes,
the invention provides the following technical scheme.
In one aspect, the invention provides the use of a C10orf71 related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf71 encoded protein in the manufacture of a product for predicting chemotherapy responsiveness or chemotherapy resistance in a patient with a tumor.
Further, the use of a C10orf 71-related molecule of the invention for the manufacture of a product for predicting chemotherapy responsiveness or chemotherapy resistance in a patient with a tumor, wherein the subject having the mutant C10orf 71-related molecule has a lower responsiveness to chemotherapy and a higher tolerance to chemotherapy, as predicted from the presence or absence of a mutation in the C10orf 71-related molecule, as compared to a subject having the wild-type C10orf 71-related molecule.
Still further, the use of a C10orf71 related molecule of the invention for the manufacture of a product for predicting chemotherapy responsiveness or chemotherapy resistance in a patient with a tumor, wherein the chemotherapy comprises treatment with one or more drugs selected from the group consisting of: platinum, nitrosourea, anti-pyrimidine, vinblastine, taxol, camptothecin, anthracycline, and antifolate.
The platinum group includes, but is not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, lobaplatin, and the like.
The nitrosoureas include, but are not limited to, carmustine, lomustine, semustine, nimustine, and the like.
The anti-pyrimidines include, but are not limited to, capecitabine, gemcitabine, ancitabine, tegafur, floxuridine, doxifluridine, eufol, etc.
The vinblastines include, but are not limited to, vinblastine, vincristine, vindesine, vinorelbine, and the like.
The taxoids include, but are not limited to, paclitaxel, docetaxel, long-acting paclitaxel, and the like.
The camptothecin includes, but is not limited to, camptothecin, irinotecan, topotecan, rubitecan, and the like.
The anthracyclines include, but are not limited to, doxorubicin, epirubicin, pirarubicin, and the like.
The antifolates include, but are not limited to, pemetrexed, loratrexed, raltitrexed, and the like.
In a particular embodiment, the chemotherapy comprises treatment with one or more drugs selected from the group consisting of: fluorouracil, capecitabine, oxaliplatin. More particularly the chemotherapy is XELOX or SOX.
In a second aspect, the present invention provides a use of a C10orf71 related molecule detection reagent for preparing a product for predicting chemotherapy responsiveness or chemotherapy tolerance of a tumor patient, wherein the C10orf71 related molecule comprises a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, and a C10orf71 encoded protein.
Further, the use of the C10orf 71-related molecule detection reagent of the present invention for the preparation of a product for predicting chemotherapy responsiveness or chemotherapy resistance of a tumor patient, wherein the subject having the mutant C10orf 71-related molecule has lower responsiveness to chemotherapy and higher tolerance to chemotherapy, as compared to a subject having a wild-type C10orf 71-related molecule, as predicted from the presence or absence of a mutation in the C10orf 71-related molecule.
Still further, the use of the C10orf 71-related molecular detection reagent of the present invention for the manufacture of a product for predicting chemotherapy responsiveness or chemotherapy resistance of a patient with a tumor, wherein the chemotherapy comprises treatment with one or more drugs selected from the group consisting of: platinum, nitrosourea, anti-pyrimidine, vinblastine, taxol, camptothecin, anthracycline, and antifolate.
The platinum group includes, but is not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, lobaplatin, and the like.
The nitrosoureas include, but are not limited to, carmustine, lomustine, semustine, nimustine, and the like.
The anti-pyrimidines include, but are not limited to, capecitabine, gemcitabine, ancitabine, tegafur, floxuridine, doxifluridine, eufol, etc.
The vinblastines include, but are not limited to, vinblastine, vincristine, vindesine, vinorelbine, and the like.
The taxoids include, but are not limited to, paclitaxel, docetaxel, long-acting paclitaxel, and the like.
The camptothecin includes, but is not limited to, camptothecin, irinotecan, topotecan, rubitecan, and the like.
The anthracyclines include, but are not limited to, doxorubicin, epirubicin, pirarubicin, and the like.
The antifolates include, but are not limited to, pemetrexed, loratrexed, raltitrexed, and the like.
In a particular embodiment, the chemotherapy comprises treatment with one or more drugs selected from the group consisting of: fluorouracil, capecitabine, oxaliplatin; more particularly the chemotherapy is XELOX or SOX.
In a third aspect, the invention provides an application of a C10orf71 related molecule in screening of drugs for treating tumors, wherein the C10orf71 related molecule comprises a C10orf71 chromosome nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, and a C10orf71 encoded protein.
Further, the use of the C10orf 71-related molecule according to the present invention for screening a drug for tumor therapy, wherein the mutant C10orf 71-related molecule, a tumor cell line having the mutant C10orf 71-related molecule, and/or an animal model having the mutant C10orf 71-related molecule are used to screen a drug having a therapeutic effect on a chemotherapy-resistant tumor.
Further, the use of the C10orf 71-related molecule according to the invention for screening of tumor treatment drugs, wherein the tumor treatment drugs include all drugs used in the prior art for treatment, further using one or more drugs selected from the group consisting of: fluorouracil, capecitabine, oxaliplatin; preferably the chemotherapy is XELOX or SOX. In a fourth aspect, the present invention provides a use of a C10orf 71-related molecule in the preparation of a cell cycle modulator, wherein the C10orf 71-related molecule comprises a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, and a C10orf 71-encoded protein.
Further, the present invention provides the use of a C10orf 71-related molecule for the preparation of a cell cycle modulator, wherein the cell cycle active state is decreased by introducing a mutation on the C10orf 71-related molecule, or increased by eliminating a mutation on the C10orf 71-related molecule.
In a fifth aspect, the invention provides a C10orf 71-related molecule, the C10orf 71-related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf 71-encoding protein.
Wherein the C10orf 71-related molecule has one or more mutations in the corresponding chromosomal DNA compared to wild type selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
Further, a C10orf71 related molecule according to the invention, wherein the C10orf71 encodes a protein having a mutation in the amino acid sequence selected from the group consisting of: mutations of D912N, R214M, L841R, C8R, and I450V.
In a sixth aspect, the present invention provides a detection reagent for the C10orf 71-related molecule, wherein the presence or absence of the above mutation in a sample to be tested can be determined by the detection reagent.
The mutation comprises one or more mutations on its chromosomal DNA selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
Still further, the above mutation results in a mutation in the amino acid sequence of the protein encoded by C10orf71 selected from the group consisting of: mutations of D912N, R214M, L841R, C8R, and I450V.
Furthermore, the detection reagent of the C10orf71 related molecule of the invention is a nucleic acid primer, a probe, an aptamer, an antibody, and the like.
In a seventh aspect, the present invention provides a use of mutations in a C10orf 71-related molecule for predicting chemotherapy responsiveness or chemotherapy tolerance in a patient with a tumor, wherein the C10orf 71-related molecule comprises a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, and a C10orf 71-encoding protein.
Further, the use of a mutation of a C10orf 71-related molecule according to the invention for predicting chemotherapy responsiveness or chemotherapy tolerance in a tumor patient, wherein the mutation of the C10orf 71-related molecule comprises one or more mutations on its chromosomal DNA selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
Still further, the use of a mutation in a C10orf 71-related molecule according to the invention for predicting chemotherapeutic responsiveness or chemotherapeutic resistance in a patient with a tumor, wherein the C10orf71 encodes a protein having a mutation in the amino acid sequence selected from the group consisting of: mutations of D912N, R214M, L841R, C8R, and I450V.
Accordingly, the invention is claimed as follows
Use of a C10orf71 related molecule in the manufacture of a product for predicting chemotherapy responsiveness or chemotherapy resistance in a patient with a tumor, said C10orf71 related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf71 encoded protein.
Use of a detection reagent for a C10orf 71-related molecule in the preparation of a product for predicting chemotherapy responsiveness or chemotherapy tolerance of a tumor patient, wherein the C10orf 71-related molecule comprises a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, and a C10orf71 encoded protein.
3. The use of any one of claims 1-2, wherein the subject having a mutant C10orf 71-related molecule is less responsive and more resistant to chemotherapy than the subject having a wild-type C10orf 71-related molecule, as predicted by the presence or absence of a mutation in the C10orf 71-related molecule.
4. The use of any one of claims 1 to 3, wherein chemotherapy comprises treatment with one or more drugs selected from the group consisting of: platinum, nitrosourea, anti-pyrimidine, vinblastine, taxol, camptothecin, anthracycline, and antifolate.
5. The use of any one of claims 1-4, wherein the platinum group includes, but is not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, lobaplatin, and the like
6. The use as claimed in any one of claims 1 to 4, wherein the nitrosoureas include, but are not limited to, carmustine, lomustine, semustine, nimustine and the like.
7. The use of any one of claims 1-4, wherein the anti-pyrimidines include, but are not limited to, capecitabine, gemcitabine, ancitabine, tegafur, floxuridine, doxifluridine, ewort, and the like.
8. The use as claimed in any one of claims 1 to 4, wherein the vinblastines include, but are not limited to, vinblastine, vincristine, vindesine, vinorelbine and the like.
9. The use of any of claims 1-4, wherein the taxoid includes, but is not limited to, paclitaxel, docetaxel, long-acting paclitaxel, and the like.
10. The use of any one of claims 1-4, wherein the camptothecin class includes, but is not limited to camptothecin, irinotecan, topotecan, rubitecan, and the like.
11. The use as claimed in any one of claims 1 to 4, wherein the anthracycline includes, but is not limited to, doxorubicin, epirubicin, pirarubicin, and the like.
12. The use of any one of claims 1 to 4, wherein the antifolates include, but are not limited to, pemetrexed, loratrexed, raltitrexed, and the like.
13. The use of any one of claims 1-4, wherein the chemotherapy comprises treatment of fluorouracil, capecitabine, oxaliplatin with one or more drugs selected from the group consisting of.
14. The use of any one of claims 1-4, wherein the chemotherapy is XeLOX or SOX.
15. The use according to any one of claims 1 to 14, wherein the tumour comprises papilloma, squamous cell carcinoma, basal cell carcinoma, adenoma or adenocarcinoma, cystadenoma or cystadenocarcinoma, adenoma multiforme, papilloma, transitional epithelium carcinoma or the like.
16. The use of claim 15, wherein the papillomas are found in the skin, nose, sinuses, throat, etc.; squamous carcinoma is found in the cervix, skin, esophagus, nasopharynx, lung, larynx, penis, etc.; basal cell carcinoma found in the head and facial skin; adenomas are found in the skin, thyroid, stomach, intestine; adenocarcinoma is found in the stomach, intestine, breast, thyroid, etc.; cystadenoma or cystadenocarcinoma is found in the ovary; pleomorphic adenomas are found in salivary glands; papilloma and transitional epithelial carcinoma are found in the bladder and renal pelvis.
17. The use of claim 15, wherein the mesenchymal tissue tumor includes, but is not limited to, fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma, lipoma, liposarcoma, leiomyoma, leiomyosarcoma, rhabdomyoma, rhabdomyosarcoma, hemangioma, lymphangioma, angiosarcoma, lymphangiosarcoma, osteoma, giant cell tumor, malignant giant cell tumor, chondrosarcoma, synovioma, synovial sarcoma, mesothelioma, malignant mesothelioma.
18. The use of claim 15, wherein the fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma are found more in the extremities; lipomas are mostly found in subcutaneous tissues, and liposarcoma is mostly found in lower limbs and behind the peritoneum; leiomyoma and leiomyosarcoma are found in the uterus and stomach intestine; rhabdomyoma and rhabdomyosarcoma are commonly found in the head and neck, genitourinary tract and extremities; hemangioma, angiosarcoma, lymphosarcoma, lymphangiosarcoma are mostly found in skin, subcutaneous tissue, tongue, lip, etc.; bone tumor, osteosarcoma and giant cell tumor are mostly found in bone tissues; chondrosarcoma is commonly seen in short bones of hands and feet, chondrosarcoma is commonly seen in pelvis, ribs, thighbone, humerus, scapula and the like; synovioma is mostly found near the joints; mesothelioma is often found in pleura, peritoneum, etc.
19. The use of claim 15, wherein the hematopoietic lymphoid tissue tumors include, but are not limited to, malignant lymphoma, various leukemias, multiple myeloma.
20. The use of claim 15, wherein the tumor of neural tissue includes, but is not limited to, neurofibroma, neurofibrosarcoma, schwannoma, malignant schwannoma, glioblastoma, meningioma, malignant meningioma, ganglionic neuroma, neuroblastoma, etc.
21. The use of claim 15, wherein said tumors of the skin and reproductive system include, but are not limited to, melanoma occurring on skin and mucosa, hydatidiform mole occurring in uterus, chorioepithelial carcinoma, malignant hydatidiform mole, supporting cell and stromal cell tumors occurring in ovary, testis, mediastinum, sacral tail, malignant supporting cell and stromal cell tumors, granulocytic tumors, malignant granulocytic tumors, seminoma, dysgerminoma, embryonal carcinoma, teratoma, malignant teratoma, and the like.
22. The use of any one of claims 1-14, wherein the tumor comprises a tumor of the respiratory system, a tumor of the digestive system, a tumor of the urinary system, a tumor of the circulatory system, a tumor of the locomotor system, a tumor of the reproductive system, other tumors, and the like. 23. The use of claim 22, wherein the respiratory tumors include, but are not limited to, lung cancer, nasopharyngeal carcinoma, laryngeal carcinoma, adenocarcinoma, and the like;
24. the use of claim 22, wherein the tumors of the digestive system include, but are not limited to, gastric cancer, liver cancer, esophageal cancer, intestinal cancer, pancreatic cancer, gallbladder cancer, peritoneal cancer, anal cancer, periampulla cancer, oral cancer, cecum cancer, parotid cancer, signet ring cell cancer, insulin cancer, etc.;
25. the use of claim 22, wherein the urological tumor includes, but is not limited to, renal cancer, bladder cancer, prostate cancer, urinary tract cancer, clear cell carcinoma;
26. the use of claim 22, wherein the tumors of the circulatory system comprise acute or chronic leukemia, lymphoma, hemangioma, aneurysm, hemangioblastoma, etc.;
27. the use of claim 22, wherein the motor system tumor comprises bone cancer, tenosynovium cancer, leiomyoma, schwannoma, and the like;
28. the use of claim 22, wherein the reproductive system tumor comprises cervical cancer, uterine cancer, ovarian cancer, testicular cancer, teratoma, choriocarcinoma, fallopian tube cancer, vaginal cancer, penile cancer, and the like;
29. the use of claim 22, wherein the other tumors comprise lipoma, breast cancer, brain tumor, squamous carcinoma, skin cancer, thyroid cancer, lip cancer, melanoma, tongue cancer, pituitary tumor, hamartoma, atheroma, glioma, sweat duct tumor, interstitial tumor, cranial tumor, sarcoma, chondroma, head and neck cancer, fibro-carcinoma, eye cancer, thoracic cancer, gum cancer, basal cell adenocarcinoma, invasive ductal carcinoma, cholesteatoma of the external auditory canal, sebaceous gland carcinoma, acoustic neuroma, pheochromocytoma, mediastinal tumor, and the like.
The use of a C10orf 71-related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orr71 exon, a C10orf71mRNA, a C10orf71 encoded protein in screening for a drug for tumor therapy 30.
31. The use of claim 30, wherein an animal model having a mutant C10orf71 related molecule, a tumor cell line having a mutant C10orf71 related molecule, and/or a mutant C10orf71 related molecule is used to screen for a drug having a therapeutic effect on a chemotherapy resistant tumor.
32. The use of any one of claims 30 to 31, wherein chemotherapy comprises treatment with one or more drugs selected from the group consisting of: platinum, nitrosourea, anti-pyrimidine, vinblastine, taxol, camptothecin, anthracycline, and antifolate.
33. The use of any one of claims 30-31, wherein the platinum group includes, but is not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, lobaplatin, and the like
34. The use as claimed in any one of claims 30 to 31, wherein the nitrosoureas include, but are not limited to, carmustine, lomustine, semustine, nimustine and the like.
35. The use of any one of claims 30-31, wherein the anti-pyrimidines include, but are not limited to, capecitabine, gemcitabine, ancitabine, tegafur, floxuridine, doxifluridine, ewort, and the like.
36. The use as claimed in any one of claims 30 to 31, wherein the vinblastines include, but are not limited to, vinblastine, vincristine, vindesine, vinorelbine and the like.
37. The use of any one of claims 30-31, wherein the taxoid includes, but is not limited to, paclitaxel, docetaxel, long-acting paclitaxel, and the like.
38. The use of any one of claims 30-31, wherein the camptothecin class includes, but is not limited to camptothecin, irinotecan, topotecan, rubitecan, and the like.
39. The use as claimed in any one of claims 30 to 31, wherein the anthracycline includes, but is not limited to, doxorubicin, epirubicin, pirarubicin, and the like.
40. The use of any one of claims 30 to 31, wherein the antifolates include, but are not limited to, pemetrexed, loratrexed, raltitrexed, and the like.
41. The use of any one of claims 30-31, wherein the chemotherapy comprises treatment of fluorouracil, capecitabine, oxaliplatin with one or more drugs selected from the group consisting of.
42. The use of any one of claims 30-31, wherein the chemotherapy is XELOX or SOX.
43. The use of any one of claims 30-42, wherein the tumor comprises papilloma, squamous cell carcinoma, basal cell carcinoma, adenoma or adenocarcinoma, cystadenoma or cystadenocarcinoma, adenoma multiforme, papilloma, transitional epithelium carcinoma, or the like.
44. The use of claim 43, wherein said papillomas are found in the skin, nose, sinuses, throat, etc.; squamous carcinoma is found in the cervix, skin, esophagus, nasopharynx, lung, larynx, penis, etc.; basal cell carcinoma found in the head and facial skin; adenomas are found in the skin, thyroid, stomach, intestine; adenocarcinoma is found in the stomach, intestine, breast, thyroid, etc.; cystadenoma or cystadenocarcinoma is found in the ovary; pleomorphic adenomas are found in salivary glands; papilloma and transitional epithelial carcinoma are found in the bladder and renal pelvis.
45. The use of claim 43, wherein said mesenchymal tissue tumor includes, but is not limited to, fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma, lipoma, liposarcoma, leiomyoma, leiomyosarcoma, rhabdomyoma, rhabdomyosarcoma, hemangioma, lymphangioma, angiosarcoma, lymphangiosarcoma, osteoma, giant cell tumor, malignant giant cell tumor, chondrosarcoma, synovioma, synovial sarcoma, mesothelioma, malignant mesothelioma.
46. The use of claim 43, wherein the fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma are found more in the extremities; lipomas are mostly found in subcutaneous tissues, and liposarcoma is mostly found in lower limbs and behind the peritoneum; leiomyoma and leiomyosarcoma are found in the uterus and stomach intestine; rhabdomyoma and rhabdomyosarcoma are commonly found in the head and neck, genitourinary tract and extremities; hemangioma, angiosarcoma, lymphosarcoma, lymphangiosarcoma are mostly found in skin, subcutaneous tissue, tongue, lip, etc.; bone tumor, osteosarcoma and giant cell tumor are mostly found in bone tissues; chondrosarcoma is commonly seen in short bones of hands and feet, chondrosarcoma is commonly seen in pelvis, ribs, thighbone, humerus, scapula and the like; synovioma is mostly found near the joints; mesothelioma is often found in pleura, peritoneum, etc.
47. The use of claim 43, wherein said hematopoietic lymphoid tissue tumors include but are not limited to malignant lymphoma, various leukemias, multiple myeloma.
48. The use of claim 43, wherein the tumor of nervous tissue includes but is not limited to neurofibroma, neurofibrosarcoma, schwannoma, malignant schwannoma, glioblastoma, medulloblastoma, meningioma, malignant meningioma, ganglionic neuroma, neuroblastoma, etc.
49. The use of claim 43, wherein said tumors of the skin and reproductive system include, but are not limited to, melanomas occurring in the skin and mucosa, hydatidiform mole occurring in the uterus, chorioepithelial cancers, malignant hydatidiform mole, supporting cell and stromal cell tumors occurring in the ovary, testis, mediastinum, sacral tail, malignant supporting cell and stromal cell tumors, granulocytic tumors, malignant granulocytic tumors, seminoma, dysgerminoma, embryonal carcinomas, teratomas, malignant teratomas, and the like.
50. The use of any one of claims 30-42, wherein the tumor comprises a tumor of the respiratory system, a tumor of the digestive system, a tumor of the urinary system, a tumor of the circulatory system, a tumor of the locomotor system, a tumor of the reproductive system, other tumors, etc.
51. The use of claim 50, wherein the respiratory tumors include, but are not limited to, lung cancer, nasopharyngeal carcinoma, laryngeal carcinoma, adenocarcinoma, and the like;
52. the use of claim 50, wherein the tumors of the digestive system include, but are not limited to, gastric cancer, liver cancer, esophageal cancer, intestinal cancer, pancreatic cancer, gallbladder cancer, peritoneal cancer, anal cancer, periampulla cancer, oral cancer, cecum cancer, parotid cancer, signet ring cell cancer, insulin cancer, etc.;
53. the use of claim 50, wherein the urological tumor includes, but is not limited to, renal cancer, bladder cancer, prostate cancer, urinary tract cancer, clear cell carcinoma;
54. the use of claim 50, wherein the tumors of the circulatory system comprise acute or chronic leukemia, lymphoma, hemangioma, aneurysm, hemangioblastoma, etc.;
55. the use of claim 50, wherein the motor system tumor comprises bone cancer, tenosynovium cancer, leiomyoma, schwannoma, and the like;
56. the use of claim 50, wherein the reproductive system tumor comprises cervical cancer, uterine cancer, ovarian cancer, testicular cancer, teratoma, choriocarcinoma, fallopian tube cancer, vaginal cancer, penile cancer, and the like.
57. The use of claim 50, wherein the other tumors comprise lipoma, breast cancer, brain tumor, squamous carcinoma, skin cancer, thyroid cancer, lip cancer, melanoma, tongue cancer, pituitary tumor, hamartoma, atheroma, glioma, sweat duct tumor, interstitial tumor, cranial tumor, sarcoma, chondroma, head and neck cancer, fibro-carcinoma, eye cancer, thoracic cancer, gum cancer, basal cell adenocarcinoma, invasive ductal carcinoma, cholesteatoma of the external auditory canal, sebaceous gland carcinoma, acoustic neuroma, pheochromocytoma, mediastinal tumor, and the like.
58.C10orf71 related molecules in the preparation of cell cycle regulators, the C10orf71 related molecules including C10orf71 chromosome nucleic acid fragment, C10orf71 exon, C10orf71mRNA, C10orf71 encoded protein.
59. The use of claim 58, wherein the cell cycle activity is decreased by introducing a mutation in a C10orf71 related molecule, or increased by eliminating a mutation in a C10orf71 related molecule.
60. The use of any one of claims 58-59, wherein the mutant chromosomal DNA on the C10orf 71-related molecule has one or more mutations selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
61. The use of any one of claims 58 to 60, wherein a mutation in a C10orf71 related molecule results in a mutation in the amino acid sequence of the protein encoded by C10orf71 selected from the group consisting of: mutations of D912N, R214M, L841R, C8R, and I450V.
62. A C10orf71 related molecule, said C10orf71 related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf71 encoded protein, wherein said C10orf71 related molecule has one or more mutations in the corresponding chromosomal DNA selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
63. The C10orf71 related molecule of claim 62, wherein the C10orf71 encoding protein has a mutation in the amino acid sequence selected from the group consisting of: mutations of D912N, R214M, L841R, C8R, and I450V.
64. The reagent for detecting a molecule associated with C10orf71 of claim 62 or 63, wherein the presence or absence of said mutation in the sample is determinable from said reagent.
65. The reagent for detecting the molecule related to C10orf71 of claim 64, which is an antibody, an aptamer, a primer, a probe, or the like.
66. The reagent of claim 64-65 for detecting the C10orf 71-related molecule, wherein the antibody is murine, yolk, human, humanized, rabbit, chimeric, heavy chain, single domain, or antigen binding fragment of an antibody.
67. A reagent for detecting molecules related to C10orf71 as claimed in claims 64-65, wherein the antibody is modified to carry enzyme label, optical label, fluorescent label, isotope label, quantum dot label, etc.
68. The reagent for detecting the molecule related to C10orf71 of claims 64-65, wherein the aptamer is natural or modified DNA, RNA, PNA, etc.
69. The reagent for detecting the molecule related to C10orf71 of claims 64-65, wherein the aptamer is single-stranded or double-stranded.
Use of a mutation in a C10orf 71-related molecule for predicting chemotherapy responsiveness or chemotherapy tolerance in a patient with a tumor, wherein the C10orf 71-related molecule comprises a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf 71-encoding protein.
71. The use of claim 70, wherein the mutation of the C10orf 71-related molecule comprises a mutation in its chromosomal DNA selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
72. The use according to claim 70 or 71, wherein the protein encoded by C10orf71 has a mutation in the amino acid sequence selected from the group consisting of: mutations of D912N, R214M, L841R, C8R, and I450V.
73. The use of any one of claims 70 to 72, wherein chemotherapy comprises treatment with one or more drugs selected from the group consisting of: platinum, nitrosourea, anti-pyrimidine, vinblastine, taxol, camptothecin, anthracycline, and antifolate.
74. The use of any one of claims 70-72, wherein the platinum group includes, but is not limited to, cisplatin, carboplatin, nedaplatin, oxaliplatin, lobaplatin, and the like
75. The use as claimed in any one of claims 70 to 72, wherein said nitrosoureas include, but are not limited to, carmustine, lomustine, semustine, nimustine and the like.
76. The use of any one of claims 70-72, wherein the anti-pyrimidines include, but are not limited to, capecitabine, gemcitabine, ancitabine, tegafur, floxuridine, doxifluridine, ewort, and the like.
77. The use as claimed in any one of claims 70 to 72, wherein the vinblastines include, but are not limited to, vinblastine, vincristine, vindesine, vinorelbine and the like.
78. The use of any one of claims 70-72, wherein the taxoid includes, but is not limited to, paclitaxel, docetaxel, long-acting paclitaxel, and the like.
79. The use of any one of claims 70-72, wherein said camptothecin includes, but is not limited to camptothecin, irinotecan, topotecan, rubitecan, and the like.
80. The use according to any one of claims 70 to 72, wherein the anthracycline includes, but is not limited to, doxorubicin, epirubicin, pirarubicin, and the like.
81. The use of any one of claims 70 to 72, wherein the antifolates include, but are not limited to, pemetrexed, loratrexed, raltitrexed, and the like.
82. The use of any one of claims 70-72, wherein the chemotherapy comprises treatment of fluorouracil, capecitabine, oxaliplatin with one or more drugs selected from the group consisting of.
83. The use of any one of claims 70-72, wherein the chemotherapy is XELOX or SOX.
84. The use of any one of claims 70-72, wherein the tumor comprises papilloma, squamous cell carcinoma, basal cell carcinoma, adenoma or adenocarcinoma, cystadenoma or cystadenocarcinoma, adenoma multiforme, papilloma, transitional epithelium carcinoma, or the like.
85. The use of claim 84, wherein said papillomas are found in the skin, nose, sinuses, larynx, etc.; squamous carcinoma is found in the cervix, skin, esophagus, nasopharynx, lung, larynx, penis, etc.; basal cell carcinoma found in the head and facial skin; adenomas are found in the skin, thyroid, stomach, intestine; adenocarcinoma is found in the stomach, intestine, breast, thyroid, etc.; cystadenoma or cystadenocarcinoma is found in the ovary; pleomorphic adenomas are found in salivary glands; papilloma and transitional epithelial carcinoma are found in the bladder and renal pelvis.
86. The use of claim 84, wherein said mesenchymal tissue tumor includes, but is not limited to, fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma, lipoma, liposarcoma, leiomyoma, leiomyosarcoma, rhabdomyoma, rhabdomyosarcoma, hemangioma, lymphangioma, angiosarcoma, lymphangiosarcoma, osteoma, giant cell tumor, malignant giant cell tumor, chondrosarcoma, synovioma, synovial sarcoma, mesothelioma, malignant mesothelioma.
87. The use of claim 84, wherein the fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma are more prevalent in the extremities; lipomas are mostly found in subcutaneous tissues, and liposarcoma is mostly found in lower limbs and behind the peritoneum; leiomyoma and leiomyosarcoma are found in the uterus and stomach intestine; rhabdomyoma and rhabdomyosarcoma are commonly found in the head and neck, genitourinary tract and extremities; hemangioma, angiosarcoma, lymphosarcoma, lymphangiosarcoma are mostly found in skin, subcutaneous tissue, tongue, lip, etc.; bone tumor, osteosarcoma and giant cell tumor are mostly found in bone tissues; chondrosarcoma is commonly seen in short bones of hands and feet, chondrosarcoma is commonly seen in pelvis, ribs, thighbone, humerus, scapula and the like; synovioma is mostly found near the joints; mesothelioma is often found in pleura, peritoneum, etc.
88. The use of claim 84, wherein the hematopoietic lymphoid tissue tumors include, but are not limited to, malignant lymphoma, various leukemias, multiple myeloma.
89. The use of claim 84, wherein the tumor of neural tissue includes, but is not limited to, neurofibroma, neurofibrosarcoma, schwannoma, malignant schwannoma, glioblastoma, malignant glioblastoma, medulloblastoma, meningioma, malignant meningioma, ganglionic neuroma, neuroblastoma, etc.
90. The use of claim 84, wherein said tumors of the skin and reproductive system include, but are not limited to, melanomas occurring in the skin and mucosa, hydatidiform mole occurring in the uterus, chorioepithelial cancers, malignant hydatidiform mole, supporting cell and stromal cell tumors occurring in the ovary, testis, mediastinum, sacral tail, malignant supporting cell and stromal cell tumors, granulocytic tumors, malignant granulocytic tumors, seminoma, dysgerminoma, embryonal carcinomas, teratomas, malignant teratomas, and the like.
91. The use of any of claims 70-72, wherein the tumor comprises a tumor of the respiratory system, a tumor of the digestive system, a tumor of the urinary system, a tumor of the circulatory system, a tumor of the locomotor system, a tumor of the reproductive system, other tumors, and the like.
92. The use of claim 91, wherein the respiratory tumor includes but is not limited to lung cancer, nasopharyngeal carcinoma, laryngeal carcinoma, adenocarcinoma, and the like;
93. the use of claim 91, wherein the tumors of the digestive system include, but are not limited to, gastric cancer, liver cancer, esophageal cancer, intestinal cancer, pancreatic cancer, gallbladder cancer, peritoneal cancer, anal cancer, periampulla cancer, oral cancer, cecum cancer, parotid cancer, signet ring cell cancer, insulin cancer, etc.;
94. the use of claim 91, wherein the urological tumor includes, but is not limited to, renal cancer, bladder cancer, prostate cancer, urinary tract cancer, clear cell carcinoma;
95. the use of claim 91, wherein the tumors of the circulatory system comprise acute or chronic leukemia, lymphoma, hemangioma, aneurysm, hemangioblastoma, etc.;
96. the use of claim 91, wherein the motor system tumor comprises bone cancer, tenosynovium cancer, leiomyoma, schwannoma, and the like;
97. the use of claim 91, wherein the reproductive system tumor comprises cervical cancer, uterine cancer, ovarian cancer, testicular cancer, teratoma, choriocarcinoma, fallopian tube cancer, vaginal cancer, penile cancer, and the like.
98. The use of claim 91, wherein the other tumors comprise lipoma, breast cancer, brain tumor, squamous carcinoma, skin cancer, thyroid cancer, lip cancer, melanoma, tongue cancer, pituitary tumor, hamartoma, atheroma, glioma, sweat duct tumor, interstitial tumor, cranial tumor, sarcoma, chondroma, head and neck cancer, fibro-carcinoma, eye cancer, thoracic cancer, gum cancer, basal cell adenocarcinoma, invasive ductal carcinoma, cholesteatoma of the external auditory canal, sebaceous gland carcinoma, acoustic neuroma, pheochromocytoma, mediastinal tumor, and the like.
Advantageous effects
The invention firstly determines the key molecular characteristics of C10orf71 gene mutation and the response of tumors to neoadjuvant chemotherapy, and based on the technical scheme of the invention, the neoadjuvant chemotherapy has the advantages of improving prognosis and determining responsive patients before the treatment starts, so that a better treatment scheme can be established. The protocol of the present invention optimizes the treatment strategy for individual GC patients as an important step.
Drawings
FIG. 1: sample collection and treatment effect assessment: (A) sample collection and omics data generation. The study included 35 GC patients who received neoadjuvant chemotherapy prior to surgery. Biopsy samples before treatment and surgically excised tumor samples after treatment were collected. Patients were divided into a responsive (n-17) and non-responsive (n-18) group based on strictly assessed radiological and pathological evidence. Multiple sets of data for pre-treatment and non-response-post-treatment samples were obtained by Whole Exome Sequencing (WES), Whole Genome Sequencing (WGS) and RNA sequencing (RNAseq). (B) Representative radiological and pathological images from responsive and non-responsive patients. Arrows indicate the lesion sites. (C) Distribution of necrosis rates in the responsive and non-responsive groups. (D) Mandard tumor regression scores in the responsive and non-responsive groups.
Figure 2 mutation characteristics in GC samples before treatment: (A) contributions from six possible substitution types in different nucleotide contexts. Relative weights (B) and microsatellite instability (MSI) scores (C) of the codinc mutant features 17 between the non-responder group and responder group. The P values are based on the Wilcoxon rank-sum test. (D) Tumor mutation burden distribution between the two groups. P values are based on a single tail t test. (B) The centerline of the- (D) box is the median, the bottom and top of the box are the first and third quartiles, and whiskers extends to the 1.5 x quartile range of the lower and upper quartiles, respectively.
Figure 3 genes with significant mutations in GC samples before treatment: (A) selected Significantly Mutated Genes (SMGs) identified by MuSiC2(FDR < 0.05) in pre-treatment tumor samples. The top and right bars show the observed mutation rate in each patient and the composition of the mutations in the selected genes, respectively. The genes are ordered by their mutation frequency, and different types of mutations are labeled with different grayscale colors. (B) C10orf71 showed significant mutation bias in the non-responsive samples (p < 0.045). The mutation sites are shown in gene cars. (C) The C10orf71 mutation was associated with resistance of the gastric cell line to cisplatin, t-test, p 1.1x10-4. (D) Cell cycle functional proteomics analysis based on 8 protein markers in a reverse phase protein array. (E) The C10orf71 mutation correlated with a lower cell cycle score in the gastric cell line, t-test, p ═ 0.015. (F) A proposed mechanistic model in which the C10orf71 mutation confers resistance to neoadjuvant chemotherapy by causing a less active cell cycle state.
Figure 4 significant somatic copy number changes in GC samples before treatment and their downstream signaling effects: (A) amplification signals for changes in somatic copy number plotted for the response versus the non-response groups. In the response group, the two oncogenes MYC and CCNE are located in unique peaks of 8q24.21 and 19q21, respectively, while the TP53 negative regulator MDM2 is in a unique peak of 12q15 in the response group. Myc (b) and MDM2(D) mRNA expression levels in the non-reactive and reactive groups. The P values are based on the single tailed Wilcoxon rank-sum test. The middle line in the box is the median, the bottom and top of the box are the first and third quartiles, and whiskers extends to the 1.5 x quartile range of the lower and upper quartiles, respectively. Enrichment of MYC target genes (C) and DNA repair pathways (E) in up-regulated genes in the reactive group relative to the non-reactive group. FDR was based on gene set enrichment analysis.
Figure 5 mutant evolution following neoadjuvant chemotherapy: (A) cancer gene mutation patterns before and after neoadjuvant chemotherapy. (B) After treatment, the top subnet is rich in mutational changes. The size of the circle represents the number of samples in the network where the mutation occurred. (C) Mutant allele frequency of the C10orf71 coding region before and after treatment in four patients. The P values are based on paired t-tests. The mutations of different patients are shown in different grey-scale colors. (D-E) schematic representation of the putative evolution of the C10orf71 mutation obtained in two patients.
FIG. 6 Gene expression and changes in tumor infiltrating immune cells following neoadjuvant chemotherapy: (A) volcano plots show genes differentially expressed between matched pre-and post-treatment samples. Important genes are shown in dark color (FC > 2, FDR < 0.05). (B) A significantly down-regulated pathway following neoadjuvant chemotherapy. Genes that were significantly differentially expressed were identified at fold change > 2 and FDR < 0.05. The bar color indicates the number of differentially expressed genes in the pathway. (C) Heat map showing fold change in mRNA expression of MYC target genes driven by treatment in MYC-amplified tumors (post-treatment/pre-treatment). Genes with significantly different expression (paired t-test, p < 0.05) were marked with light (down-regulation) and dark (up-regulation). (D) Differential expression of GC therapeutic targets in pre-and post-treatment samples. P values were calculated based on paired Wilcoxon rank-sum. (E) Fraction of neutrophils and dendritic cells in the sample before and after treatment. P values were calculated based on paired t-tests. (D) And (E) the middle line of the box is the median, the bottom and top of the box are the first and third quartiles, and whiskers extends to the 1.5 x quartile range of the lower and upper quartiles, respectively.
Detailed Description
The present invention is described in more detail below to facilitate an understanding of the present invention.
Defining:
tumor (tumor) refers to a new organism formed by local histiocyte proliferation under the action of various tumorigenic factors. Tumors include both benign and malignant tumors, depending on their cellular characteristics and the extent of damage to the body.
Tumors include epithelial tumors, mesenchymal tumors, lymphohematopoietic tumors, neural tumors, skin and reproductive tumors, etc., depending on the source of the tumorigenesis.
The epithelial tumors include, but are not limited to, papilloma, squamous cell carcinoma, basal cell carcinoma, adenoma or adenocarcinoma, cystadenoma or cystadenocarcinoma, pleomorphic adenoma, papilloma, transitional epithelial carcinoma, and the like.
Wherein papilloma is found on skin, nose, sinuses, throat, etc.; squamous carcinoma is found in the cervix, skin, esophagus, nasopharynx, lung, larynx, penis, etc.; basal cell carcinoma found in the head and facial skin; adenomas are found in the skin, thyroid, stomach, intestine; adenocarcinoma is found in the stomach, intestine, breast, thyroid, etc.; cystadenoma or cystadenocarcinoma is found in the ovary; pleomorphic adenomas are found in salivary glands; papilloma and transitional epithelial carcinoma are found in the bladder and renal pelvis.
The mesenchymal tissue tumor includes, but is not limited to, fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma, lipoma, liposarcoma, leiomyoma, leiomyosarcoma, rhabdomyoma, rhabdomyosarcoma, hemangioma, lymphangioma, angiosarcoma, lymphangosarcoma, osteoma, osteosarcoma, giant cell tumor, malignant giant cell tumor, chondrosarcoma, synovioma, synovial sarcoma, mesothelioma, malignant mesothelioma.
Among them, fibroma, fibrosarcoma, benign or malignant fibrous histiocytoma are often found in the extremities; lipomas are mostly found in subcutaneous tissues, and liposarcoma is mostly found in lower limbs and behind the peritoneum; leiomyoma and leiomyosarcoma are found in the uterus and stomach intestine; rhabdomyoma and rhabdomyosarcoma are commonly found in the head and neck, genitourinary tract and extremities; hemangioma, angiosarcoma, lymphosarcoma, lymphangiosarcoma are mostly found in skin, subcutaneous tissue, tongue, lip, etc.; bone tumor, osteosarcoma and giant cell tumor are mostly found in bone tissues; chondrosarcoma is commonly seen in short bones of hands and feet, chondrosarcoma is commonly seen in pelvis, ribs, thighbone, humerus, scapula and the like; synovioma is mostly found near the joints; mesothelioma is often found in pleura, peritoneum, etc.
Such hematopoietic lymphoid tissue tumors include, but are not limited to, malignant lymphoma, various leukemias, multiple myeloma.
The neural tissue tumor includes, but is not limited to, neurofibroma, neurofibrosarcoma, schwannoma, malignant schwannoma, glioblastoma, malignant glioma, medulloblastoma, meningioma, malignant meningioma, ganglionic neuroma, neuroblastoma, etc.
Such tumors of the skin and reproductive system include, but are not limited to, melanoma that occurs on the skin and mucosa, hydatidiform mole that occurs in the uterus, chorioepithelioma, malignant hydatidiform mole, supporting cell and stromal cell tumors that occur in the ovary, testis, mediastinum, sacral tail, malignant supporting cell and stromal cell tumors, granulocytic tumors, malignant granulocytic tumors, seminoma, dysgerminoma, embryonal carcinoma, teratoma, malignant teratoma, and the like.
According to the tumor and the occurring part, the tumor can be divided into respiratory system tumor, digestive system tumor, urinary system tumor, circulatory system tumor, motion system tumor, reproductive system tumor, other tumors, etc.
The respiratory system tumors include but are not limited to lung cancer, nasopharyngeal carcinoma, laryngeal carcinoma, adenocarcinoma and the like;
the digestive system tumors include but are not limited to gastric cancer, liver cancer, esophageal cancer, intestinal cancer, pancreatic cancer, gallbladder cancer, peritoneal cancer, anal cancer, periampulla cancer, oral cancer, cecum cancer, parotid gland cancer, signet ring cell cancer, insulin cancer and the like;
the urological tumors include, but are not limited to, renal cancer, bladder cancer, prostate cancer, urinary tract cancer, clear cell carcinoma;
the circulatory system tumor comprises acute or chronic leukemia, lymphoma, hemangioma, aneurysm, hemangioblastoma and the like;
the motor system tumor comprises bone cancer, ganglion cancer, leiomyoma, schwannoma and the like;
the reproductive system tumor comprises cervical cancer, uterine cancer, ovarian cancer, testicular cancer, teratoma, choriocarcinoma, fallopian tube cancer, vaginal cancer, penile cancer, etc.;
the other tumors include lipoma, breast cancer, brain tumor, squamous carcinoma, skin cancer, thyroid cancer, lip cancer, melanoma, tongue cancer, pituitary tumor, hamartoma, atheroma, glioma, syringoma, interstitial tumor, cranial tumor, sarcoma, chondroma, head and neck cancer, fibrocarcinoma, eye cancer, thoracic cancer, gum cancer, basal cell adenocarcinoma, invasive ductal carcinoma, cholesteatoma of the external auditory canal, sebaceous gland carcinoma, acoustic neuroma, pheochromocytoma, mediastinal tumor, and the like.
Mutation (mutation) refers to an alteration in the nucleotide sequence of an organism, virus or extrachromosomal DNA genome. Naturally, mutations are caused by errors in DNA replication (particularly meiosis) or by repair of errors following DNA damage (e.g., exposure to radiation or carcinogens).
Small-scale mutations affect one or a few nucleotides in a gene, with point mutations affecting only one nucleotide being referred to as point mutations. Small-scale mutations include: insertions, deletions, substitutions, and the like.
An insertion is the addition of one or more additional nucleotides to the DNA.
A deletion is the removal of one or more nucleotides from the DNA.
Substitutions are typically made by chemical or DNA replication aberrations that replace a single nucleotide in a gene with another nucleotide.
Point mutations are mutations of a single base pair in a gene, and point mutations occurring in the protein-coding region of a gene can be classified into synonymous mutations and nonsynonymous mutations (missense mutations).
Large-scale mutations involve mutations in chromosomal structures, including: amplification, deletion, rearrangement, and the like.
Amplification results in an increase in copy number within each region of the chromosome, thereby increasing the number of genes in the chromosome.
Deletions are large segments of chromosomal deletions resulting in loss of genes in this region.
Rearrangements include chromosomal translocations (exchange of part of the genetic material for non-homologous chromosomes), chromosomal inversions (inversion of the orientation of chromosomal segments), non-homologous crossovers, interstitial deletions (removal of a stretch of DNA from a single chromosome causes an intrachromosomal deletion), heterozygous deletions (loss of one allele by deletion or recombination in an organism with two different alleles), and the like.
Antibodies (antibodies) are proteins which are produced by the body as a result of antigenic stimulation and which have a protective effect and are large proteins which are secreted by plasma cells and which are used by the immune system to recognize and neutralize foreign substances. The nature of antibodies is a class of immunoglobulins that specifically bind to antigens, and antibodies generally include five classes, IgG, IgM, IgA, IgD, and IgE, and IgY in the yolk of avians.
The antibody in a broad sense includes not only a natural antibody but also various forms of antibody derivatives such as a humanized antibody, a chimeric antibody, a single-chain antibody, a monovalent antibody, Fab, F (ab)2, a heavy-chain antibody, and a single-domain antibody.
Aptamers (aptamers) are oligonucleotide sequences that are repeatedly screened from random oligonucleotide sequence libraries synthesized in vitro by the artificial technology of systematic evolution of ligands by exponential enrichment (SELEX) and can be combined with target molecules with extremely high affinity and specificity. Aptamers may be DNA, RNA, PNA, etc., either single-stranded or double-stranded. The aptamer has the advantages of wide target molecule range, high affinity with the ligand, strong specificity, high stability, simple preparation and the like.
A Primer (Primer) is a macromolecule having a specific nucleotide sequence, which is stimulated to synthesize at the start of nucleotide polymerization, and is covalently linked to a reactant. The primers are typically two oligonucleotide sequences synthesized by man, one complementary to one DNA template strand at one end of the target region and the other complementary to the other DNA template strand at the other end of the target region, and function as a starting point for nucleotide polymerization, from which 3' end the nucleic acid polymerase can synthesize a new nucleic acid strand. Primers designed artificially in vitro are widely used for polymerase chain reaction, sequencing, probe synthesis, and the like.
Probes (probes) are single-stranded nucleic acid molecules that bind complementarily to a target nucleotide sequence. Generally, probes can be classified into specific DNA probes, specific RNA probes, and synthetic oligonucleotide probes, depending on the source of the probe.
Example (b): verification of C10orf 71-related molecule responsiveness to chemotherapy in patients with GC tumors
Patient recruitment and sample cohort
GC patients studied in this invention (> T2N + M0, UICC-AJCC version 8) were recruited in the university of beijing tumor hospital (beijing, china) in 2015 to 2018. The study was performed according to the declaration of helsinki and was approved by the ethical committee of the cancer hospital, beijing university (IRB approval No., 2019KT 05). All patients provided written informed consent prior to treatment, sample collection and analysis. Tumors were collected by biopsy and matched to adjacent non-tumor tissue prior to neoadjuvant therapy. For patients with no adjacent non-tumor tissue available, blood samples were taken for replacement. All patients then received a fluorouracil-based capecitabine/S-1 + oxaliplatin treatment regimen (XELOX [ oxaliplatin, 130mg/m2, intravenous, day 1; capecitabine, 1000mg/m2, oral, day 1 to 14 ] or SOX [ oxaliplatin 130mg/m2, day 1 intravenous drip; S-1, 40-60mg, 2 times daily, oral, 2-4 cycles from day 1 to 14, and evaluated the response to treatment before surgery, pathological characteristics were evaluated according to the madard TRG score, which was performed by three independent blind pathologists (lymart, liu, and zhang) on all patients according to the standard. Cancer 73, 2680-2686(1994). To reduce classification noise, only three pathologists were focused on consensus assessment patients, and finally 35 patients (17 responses and 18 non-responses) were included as a study cohort. Tumor samples were further collected post-operatively. For patients who responded, no post-treatment tumor samples were collected.
Multi-set mathematical data generation
For 35 GC patients, genomic DNA in tissue (or blood) samples was extracted using AllPrep DNA Mini Kit (QIAGEN) and QIAamp DNA blood Mini Kit (QIAGEN), respectively. For whole exome sequencing, 1. mu.g of DNA was cut into short fragments (150-250bp) using Bioruptor (Diagenode). Repairing the resulting DNA fragment. Adaptor fragments are then ligated to both ends of the fragment. DNA fragments of the target size are selected. Thereafter, Polymerase Chain Reaction (PCR) was performed, and the resulting mixture was purified. Exome capture was performed using SureSelect Human All Exon V6(Agilent) according to the manufacturer's protocol. The hybridized mixture was then amplified by PCR. The validated DNA library was then sequenced on Illumina NovaSeq 6000. For whole genome sequencing, 1. mu.g of genomic DNA was sheared to 150-250bp using Bioruptor (Diagenode). The DNA library was then generated using the standard protocol of Truseq nano dnakit (illumina). Paired-end sequencing (paired-end runs) was performed using Illumina NovaSeq 6000, and the library was sequenced to a minimum depth of 6 × base coverage. For RNA sequencing, total RNA was extracted from fresh tissue using the AllPrep RNA mini kit (QIAGEN). For each sample, a library was generated by TruSeq RNA v2 kit (illumina) using 3 μ g total RNA. The library was sequenced with Illumina NovaSeq 6000 and an average of 3,300 million 2 x 150bp paired ends per sample.
Analysis of variant data
Full exome sequencing read pairs were trimmed and the remaining read pairs had < 3% N bases and > 50% high quality bases. The resulting high quality reads were aligned to the human reference genome (Homo _ sapiens _ assembly19) using Burrows-Wheeler Aligner 0.7.17 (h.li, r.durbin, Fast and acid short read alignment with Burrows-Wheeler transform. bioinformatics 25, 1754-. To improve The alignment accuracy, The BAM file was processed using a Genome Analysis Toolkit (version 3.8.1) (a. mckenna et al, The Genome Analysis Toolkit: a MapReduce frame for analyzing next-generation DNA sequencing data. Genome Res 20, 1297-1303(2010)) by: repeat entries are labeled, local re-alignments are performed around high confidence insertions and deletions and base quality is re-aligned. Based on about 7,000 high frequency SNP sites, matched pre-treatment, post-treatment and normal samples from the same patient were confirmed. A variant Calling pipeline developed in the cancer Genomic map MC3 project was used to identify high confidence somatic base substitutions and insertions/deletions (K.Ellrott et al, Scalable Open Science Approach for Mutation Calling of Multiple Genomic pipelines. cell Syst 6, 271-281 e277 (2018)). In short, this pipeline uses six callers to invoke the substitution mutation and three callers to identify the insertion/deletion with detailed annotation information. Only at least two caller-supported substitution mutations and insertions/deletions were retained for further analysis. WES somatic base substitutions were further validated based on RNA-seq data from the same samples. An alignment was generated using TopHat2 (D.Kim et al, TopHat 2: acquisition alignment of transformations in the presentation of insertions, deletions and gene fusions. genome Biol 14, R36 (2013)). For each substitution site, the coverage and number of mutation reads were calculated from RNA-seq BAM files from the same sample. For sites with sufficient RNA-seq coverage (. gtoreq.10X), sites with at least two reads can be considered to have validated the base substitution of interest. The characteristics of mutations in the pre-treatment samples were determined using R package deconstructed Sigs v1.8.0 as a matrix of mutation characteristics based on COSMIC characteristics (R.Rosenthal, N.McGranahan, J.Herreo, B.S.Taylor, C.Swanton, deconstructed Sigs: delinating biological processes in single structures differentiation DNA repair details and patterns of cancer evolution.genome Biol 17, 31 (2016)). The Microsatellite instability status of each tumor was assessed using MANTIS (E.A. Kautto et al, Performance evaluation for Rapid detection of pan-cancer specificity with MANTIS. on target 8, 7452-1197463 (2017)), and 2539 loci (S.J. Salipate, S.M. Scrogins, H.L. Hampel, E.H. Turner, C.C. Pritch, Microcuterite specificity detection sequencing. clin chemistry m 60, 1192-1199 (2014)) obtained from the mSINGS package were used for this analysis. The apparently mutated genes were identified in all pre-treatment samples using MuSiC2(18.N.D. Des et al, MuSiC: identifying mutation in cancer genes. genome Res 22, 1589-1598 (2012)). According to the Fisher's exact test, Maftools (A. Mayakonda, D.C.Lin, Y.Assenov, C.P1ass, H.P.Koeffler, Maftools: effective and comprehensive analysis of physiological variables in cancer. genome Res 28, 1747-1756(2018)) were used to explore SMGs that showed clear mutational bias between non-responsive and responsive samples. To detect the gene group rich in the base substitution change after the treatment, HotNet2 (29) was used.
M.D. Leiserson et al, Pan-cancer network analysis identities combinations of rare acidic pathologies and proteins Nat Genet 47, 106-114(2015), identified apparently mutated sub-networks based on the caloric value of each protein. Only genes with altered events were retained. An altered event is defined as a different non-silent variation of the same patient before and after treatment. The heat score is limited to major drivers or genes with at least two events of change. The consensus sub-network is visualized using the HINT + HI2012, MultiNet and iRefIndex interactive networks. Fisher's exact test was used to determine statistical significance.
Drug response determination and analysis
Cisplatin response data (AUC score) were obtained from GDSC for 21 gastric Cancer cell lines with CCLE mutation profile data (W. Yang et al, Genomics of Drug Sensitivity in Cancer (GDSC): a resource for thermal biological discovery in Cancer cells. nucleic Acids Res 41, D955-961 (2013)). In cell lines, 3 contained non-silent single nucleotide variations in C10orf71, while 18 were wild-type. The Student's t test was used to assess the difference between cell lines with the C10orf71 mutation and cell lines with the wild type.
Reverse phase protein array analysis
For the RPPA experiments, 18 gastric lines were first confirmed by short tandem repeats and then prepared. The antibody was verified as previously described (M.Ghandi et al, Next-generation Characterization of the Cancer Cell Line encyclopedia. Nature 569.503-508 (2019); J.Li et al, Characterization of Human Cancer Cell Lines by Reverse-phase Protein arrays. Cancer Cell 31, 225-239 (2017)). As previously described, RPPA data was generated by the RPPA core agency of the MD anderson cancer center. RPPA sections were first quantified using arraypro (meda cybernetics) to generate signal intensity, then processed by SuperCurve to estimate relative protein expression levels, and then normalized by median modification. The RPPA slide quality was evaluated by a quality control classifier, and only those slides above 0.8 (range: 0-1) were retained for further analysis. Protein markers were divided into 11 signal pathways and pathway scores were calculated based on the orientation of the protein members. The Student's t test was used to assess the difference between the cell line with the C10orf71 mutation and the wild type cell line, and FDR was used for multiple test corrections.
Somatic copy number Change analysis
Whole genome sequencing data was used to infer copy number values in samples before and after treatment. The whole exome sequencing read pairs were trimmed according to the criteria used for WES data. Reads were aligned to the human reference genome (Homo _ sapiens _ assombly 19) using BWA 0.7.17. SCNAs were then estimated by Control-FREEC (version 11.5) based on matching normal tumor pairs with a window size of 5kb (v. boeva et al, Control-FREEC: a tool for assessing copy number and adaptive content using next-generation sequencing data. bioinformatics 28, 423-425 (2012)). Regions with statistically significant copy number variation frequencies were identified using gist 2.0(c.h. memel et al, gist 2.0 defects sensitive and reliable localization of the targets of local acidic copy-number alteration in human cameras Biol 12, R41(2011)) based on the corresponding segmentation values. The amplification and deletion thresholds were set at 0.1. Using the same tools, parallel analysis was performed on whole exome sequencing data as an independent dataset to verify the SCNA peaks identified by whole genome sequencing.
Clonal structure analysis of sample pre-and post-treatment
To explore the new adjuvant chemotherapy-driven clonal evolution, dynamic changes in clonal structure were inferred using sciClone (c.a. miller et al, sciClone: inducing cyclic architecture and tracking the activity and temporal patterns of tumor evolution. plos computer Biol 10, e1003665 (2014)). Copy number values based on whole exome sequencing data inferred by Control-FREEC were used to exclude SNVs from the copy number change region for clonal analysis. The inferred subclone evolution was further verified using another tool, PyClone (A.Roth et al, PyClone: statistical information of clinical position structure in cancer. Nat Methods 11, 396-398 (2014)).
Analysis of Gene expression
Gene expression was quantified based on RNA sequencing data using the alignment-free tool Kallisto (N.L.Bray, H.Pimentel, P.Melsted, L.Pachter, Near-optimal genomic RNA-seq quantification. Nat Biotechnol 34, 525-527 (2016)). The output of Kallisto was converted to gene-level counts and TPM (c. soneson, m.i.love, m.d. robinson, Differential analysis for RNA-seq: transcript-level assays. f1000res 4, 1521(2015)) using txiprort. To identify genes that are differentially expressed between the responsive and non-responsive groups, a t-test was performed on each gene and they were ranked according to t-stability. The hallmark pathway of enrichment was determined using a pre-rank based Gene set enrichment assay (A. Subramanian et al, Gene set expression analysis: a knowledge-based approach for expressing genome-wide expression profiles. Proc Natl Acad Sci U S102, 15545 and 15550 (2005)). To identify genes differentially expressed between tumor samples at different treatment stages in non-responders, a multifactorial model was fitted with patient ID as blocking factor, and then a Wald test was performed for treatment stage effects using DESeq2 (m.i. love, w.huber, s.anders, modeled evaluation of fold change and dispersion for RNA-seq data with DESeq2.genome Biol 15, 550 (2014)). Genes with FDR < 0.05 and fold change > 2.0 or < 0.5 were selected as differentially expressed genes. Overexpression analysis of the MSigDB marker Gene set achieved by cluster Profile (G.Yu, L.G.Wang, Y.Han, Q.Y.He, cluster Profile: an R package for formulating biological enzymes systems clusters OMICS 16, 284 287(2012)) was performed separately for genes up-regulated and down-regulated in the post-treated samples (E.I.Boyle et al, GO:. TermFinder- -open source software for accessing Gene on-regulated and designing characterized genes associated with a Gene of genes. Bioinformatics 20, 3710- -. The differential gene expression of MYC target genes before and after MYC-amplified non-responder treatment was further compared using Student's t-test. Tumor infiltrating immune cell abundance was inferred from mRNA expression data using TIMERs (b.li et al, Comprehensive analyses of tumor immunity: modalities for cancer immunity. genome biology 17, 174 (2016)). Paired Student's t-test was performed to identify differences in cellular composition between matching pre-and post-treatment samples.
Biopsy tumor samples from 35 pre-neoadjuvant-chemotherapy GC patients were subjected to multicohort sequencing (detailed patient and sample information is shown in fig. 1A). For these cases, whole exome sequencing was performed on tumor samples as well as matched germline DNA samples, which were used primarily to identify somatic base substitutions and small insertions/deletions. In 32 of 35 cases (3 excluded due to insufficient DNA quantity), whole genome sequencing was also performed, as well as matched germline samples for identification of somatic copy number changes (SCNA). In addition, RNA sequencing was performed on all tumor samples to characterize the mRNA expression profile of the protein-encoding genes. Following biopsy, patients received neoadjuvant chemotherapy based on 5-fluorouracil + oxaliplatin for 2-4 cycles. Cases were then divided into two categories based on a strict assessment of radiology and pathology evidence by three independent pathologists: response (n-17) and non-response (n-18). Representative radiological and pathological images are shown in fig. 1B. In the response group, the necrosis rate was higher than 65% in all cases, while in the non-response group, the necrosis rate was lower than 15% (fig. 1C). The Mandard tumor regression rating was < 2 for the response group cases, 4 or 5 for the non-response group cases (FIG. 1D). Typically, patients in the response group were younger than those in the non-response group (Wilcoxon rank-sum test, 17: 18 for non-response and non-response, 0.047 for p); patients in the non-responsive group tend to be advanced (Wilcoxon rank-sum test, 17: 18 for non-responsive and non-responsive, and 0.023 for p).
After receiving neoadjuvant chemotherapy in 18 patients in the non-responsive group, fresh tumor samples were obtained surgically from 14 of these and these post-treatment samples were subjected to whole exome sequencing (fig. 1A). For these cases, whole genome sequencing (for 13 cases) and RNA sequencing were also performed, respectively. In contrast, for the patients in the response group, tumor tissue was not obtained due to lesions with low tumor cell content resulting from good response to neoadjuvant chemotherapy. In summary, this experimental design enables the identification of a variety of molecular aberrations (bulk base substitutions SCNA and gene expression) involving tumor response to neoadjuvant chemotherapy from different perspectives (response versus non-response, pre-and post-treatment).
The composition of six possible base pair substitutions was examined and it was found that the T > G substitutions showed the most pronounced pattern between the responsive and non-responsive groups, especially when the substitution sites flank C and T (FIG. 2A). Consistently, in decomposing the mutation profile into different mutation signatures, the COSMIC signature 17(T > G) (11.l.b. alexandrov et al, Clock-like biological processes in human acidic cells. nat gene 47, 1402-1407 (2015)) is a signature previously observed in esophagus and stomach cancer and related to byproducts of oxidative damage (12.m.tomkova, j.tomek, s.kriustiis, b.schuster-bocker, mutional distribution with DNA replication time and expression biology. genome 19, 129 (2018)). The response group contributed much higher than the non-response group (Wilcoxon rank-sum test, p 0.049, response vs non-response 17: 18, fig. 2B). Next, checkDistribution of microsatellite instability (MSI) scores in these tumors calculated by MANTIS was calculated (13.E.A. Kautto et al, Performance evaluation for rapid detection of pan-cancer microsalt specificity with MANTIS. Oncostatt 8, 7452-7463 (2017)). Surprisingly, MSI scores were found to be significantly higher in the non-responsive group than in the responsive group (Wilcoxon rank-sum test, p 0.022, non-responsive vs. 17: 18, fig. 2C). Thus, the non-responsive group was tested for the presence of higher mutation burden and confirmed this pattern (Student's t test, non-response vs non-response 17: 18, p 0.04, fig. 2D and fig. 3A). Preclinical data indicate that MSI-H status colorectal tumors are resistant to 5-fluorouracil-based chemotherapy (m.hewind, c.j.Lord, S.a.Martin, D.cunningham, A.ashworth, Mismatch repair specific tumor cancer in the same of transgenic tumor # nat Rev clean Oncol 7,
Figure BDA0002273721740000271
(2010) (ii) a M.Koopman et al, Predictive and qualitative markers for the outome of therapy in advanced clinical Cancer, a retroactive analysis of the phase III random CAIRO studyr, Eur J Cancer 45, 1999 + 2006(2009). Clinically, only patients with MSI-negative colorectal Cancer benefit, while MSI-H Does not benefit (16.C.M. Ribic et al, Tumor microsatellite-instabilization status as a predictor of recent from fluorogenic-based adjuvant chemotherapy for colon Cancer. N Engl J Med, 247-349 (2003)), which makes MSI-H status a powerful predictor of non-response to 5-fluorouracil-based chemotherapy (G.Des Guetz et al, Does microsatellite occlusion prediction of the efficacy of clinical study in clinical fashion review with a parameter of evaluation. Eur J.45, 11896 (2009)). This result provides the first evidence that MSI-H status can also be used as a predictor of non-response to neoadjuvant chemotherapy in GC patients.
To identify individual mutant genes that may play a role in influencing therapeutic response, MuSiC2(18.N.D. Dees et al, MuSiC: identifying mutant amino acid si) was next usedgenome Res 22, 1589-1598(2012), identified Significantly Mutated Genes (SMG) (p 2.9 × 10)-4FDR ═ 0.05). The most mutated genes included TP53, P13KCA, RNF43, ARIDA1 and KRAS as previously reported in other gastric cancer cohorts (19.N. cancer Genome Atlas Research, Comprehensive molecular characterization of organic Adenocercinoma. Nature 513, 202-209(2014)) (FIG. 3A). Among the SMGs identified in this and previous studies (19.n. cancer Genome Atlas research. comprehensive molecular characterization of biological adonnarity. nature 513, 202-209(2014)), C10orf71 is the only gene that shows a clear pattern between the responsive and non-responsive groups: mutations were found in all 5 non-responsive samples, but none (Fisher exact test, p 0.04, FDR < 0.25) and no recurrent mutations were detected (fig. 3B). To validate this observation, based on drug response data using a panel of 21 gastric cancer cells (20), it was indeed observed that cell lines with the C10orf71 mutation were more resistant to cisplatin (the equivalent platinum drug included in the neoadjuvant chemotherapy regimen) than these wild-type cell lines (Student's t test, nmut vs.nWT=3∶18,p=1.1*10-4Fig. 3C). To gain more mechanistic insight, functional proteomic data of gastric cell lines were analyzed using reverse phase protein arrays. Cell lines with the C10orf71 mutation were found to show significantly lower cell cycle scores (based on 8 protein markers) than those of the wild type (Student's t test, n)mut vs.nWT3: 18, p 0.015, fig. 3D, E). As cell cycle arrest is the major mechanism of action of platinum-based drugs, without wishing to be bound by theory, the inventors speculate that the C10orf71 mutation therein confers resistance to neoadjuvant chemotherapy by causing a less active cell cycle state (fig. 3F). These results indicate that mutations in this gene are potential biomarkers of resistance to neoadjuvant chemotherapy.
Major somatic base substitution changes following neoadjuvant chemotherapy
Genomic evolution of GC under neoadjuvant chemotherapy was examined using parallel whole exome sequencing data of 14 matched pre-and post-treatment sample pairs (mean coverage > 200). Overall, no significant change in mutation burden or mutation characteristics was found. For the known cancer drivers, some tumors showed critical mutational changes (loss or gain of mutation) before and after treatment, while other tumors showed the same mutational signature (fig. 5A). To systematically identify genes or pathways that exhibit the most frequently occurring mutational changes, HotNet2 was used to search for protein interaction networks that are rich in such signals (29.M.D. Leiserson et al, Pan-cancer network analysis identities associations and protein complexes Nat Gene 47, 106-. One top subnet identified consisted of IRS1, IRS2, PIK3CA, JAK1 and IL6ST (fig. 5B). In particular, IRS1 plays a key role in signaling insulin and insulin-like growth factor-1 (IGF-1) receptors to the PI3K/AKT pathway, which continually acquired new mutations in five post-treatment samples (Fisher's exact test, p 0.041), suggesting the existence of a recurrent resistance mechanism. Analysis of responsive and non-responsive pretreatment samples indicated that mutations in C10orf71 contribute to improved treatment resistance. If so, it would be desirable to increase the frequency of allelic mutations or obtain new mutations in the post-treatment sample, since tumor cells that are susceptible to chemotherapy (i.e., those tumor cells that do not have such mutations) would likely be removed by selection during the course of treatment. Of the 14 pairs of samples investigated, 4 showed a C10orf71 mutation in the pre-or post-treatment samples, two of which acquired a new mutation after neoadjuvant chemotherapy. A significant increase in mutant allele frequency was indeed observed over the five mutation sites detected compared to the pre-treatment samples (figure 5C, Student's t test, n-8 pairs, p < 0.05; the same pattern was true even after tumor purity was adjusted). Fig. 5D and 5E show schematic diagrams of the putative evolution of the two cases, in which the obtained C10orf71 mutation gradually increased its allele frequency. Although the true evolutionary trajectory is difficult to validate, this result suggests that the C10orf71 mutation has a key role in influencing tumor response.
Figure BDA0002273721740000301
The foregoing describes preferred embodiments of the present invention, but is not intended to limit the invention thereto. Modifications and variations of the embodiments disclosed herein may be made by those skilled in the art without departing from the scope and spirit of the invention.

Claims (10)

  1. Use of a C10orf71 related molecule in the manufacture of a product for predicting chemotherapy responsiveness or chemotherapy tolerance in a patient with a tumor, the C10orf71 related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf71 encoding protein.
  2. Use of a detection reagent for a C10orf 71-related molecule in the preparation of a product for predicting chemotherapy responsiveness or chemotherapy tolerance of a tumor patient, wherein the C10orf 71-related molecule comprises a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, and a C10orf71 encoded protein.
  3. The application of C10orf71 related molecules in screening tumor treatment medicines, wherein the C10orf71 related molecules comprise C10orf71 chromosome nucleic acid fragments, C10orf71 exons, C10orf71mRNA and C10orf71 encoding proteins.
  4. 4. The use of claim 3, wherein an animal model having a mutant C10orf71 related molecule, a tumor cell line having a mutant C10orf71 related molecule, and/or a mutant C10orf71 related molecule is used to screen for a drug having a therapeutic effect on a chemotherapy resistant tumor.
  5. Use of a C10orf 71-related molecule in the preparation of a cell cycle modulator, said C10orf 71-related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf 71-encoding protein.
  6. 6. Use according to claim 5, wherein the cell cycle activity is reduced by introducing a mutation in a C10orf71 related molecule or increased by eliminating a mutation in a C10orf71 related molecule.
  7. 7. A C10orf71 related molecule, said C10orf71 related molecule comprising a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf71 encoded protein, wherein said C10orf71 related molecule has one or more mutations in the corresponding chromosomal DNA selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
  8. 8. The C10orf71 related molecule of claim 7, wherein the C10orf71 encoding protein has a mutation in the amino acid sequence selected from the group consisting of: mutations of D912N, R214M, L841R, C8R, and I450V.
  9. Use of a mutation in a C10orf 71-related molecule for predicting chemotherapy responsiveness or chemotherapy tolerance in a patient with a tumor, wherein the C10orf 71-related molecule comprises a C10orf71 chromosomal nucleic acid fragment, a C10orf71 exon, a C10orf71mRNA, a C10orf 71-encoding protein.
  10. 10. The use of claim 9, wherein the mutation of the C10orf 71-related molecule comprises having one or more mutations on its chromosomal DNA selected from the group consisting of: a G → A mutation at position 50533324, a G → T mutation at position 50531231, a T → G mutation at position 50533112, a T → C mutation at position 50530612, and an A → G mutation at position 50531938 of chromosome 10.
CN201911117303.0A 2019-11-14 2019-11-14 Tumor marker and application thereof Active CN112795647B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201911117303.0A CN112795647B (en) 2019-11-14 2019-11-14 Tumor marker and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201911117303.0A CN112795647B (en) 2019-11-14 2019-11-14 Tumor marker and application thereof

Publications (2)

Publication Number Publication Date
CN112795647A true CN112795647A (en) 2021-05-14
CN112795647B CN112795647B (en) 2022-08-16

Family

ID=75803942

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911117303.0A Active CN112795647B (en) 2019-11-14 2019-11-14 Tumor marker and application thereof

Country Status (1)

Country Link
CN (1) CN112795647B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110607371A (en) * 2019-10-14 2019-12-24 普瑞基准科技(北京)有限公司 Stomach cancer marker and application thereof
CN114525344A (en) * 2022-04-22 2022-05-24 普瑞基准科技(北京)有限公司 Kit for detecting or assisting in detecting tumor-related gene variation and application thereof
CN114561472A (en) * 2022-04-27 2022-05-31 普瑞基准科技(北京)有限公司 Kit for detecting or assisting in detecting tumor-related gene variation and application thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105916876A (en) * 2013-09-16 2016-08-31 分子医学研究中心责任有限公司 Mutant calreticulin for the diagnosis of myeloid malignancies
CN105999250A (en) * 2011-05-24 2016-10-12 生物技术公司 Individualized vaccines for cancer
CN107148481A (en) * 2014-07-14 2017-09-08 苏黎世大学医学与自然科学院 The patient for suffering from BRAF positive cancers for determination is BRAF inhibitor non-response persons and is the tool and method of MAPK/ERK inhibitor respondents
KR20180029776A (en) * 2016-09-13 2018-03-21 경희대학교 산학협력단 A instrument on pattern identification for lung cancer using gene sequencing
CN107944224A (en) * 2017-12-06 2018-04-20 懿奈(上海)生物科技有限公司 Build method and the application of skin-related gene standard type database
JP2018143178A (en) * 2017-03-06 2018-09-20 国立大学法人三重大学 Genetic risk detection method for cardiovascular disease
CN110607371A (en) * 2019-10-14 2019-12-24 普瑞基准科技(北京)有限公司 Stomach cancer marker and application thereof

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105999250A (en) * 2011-05-24 2016-10-12 生物技术公司 Individualized vaccines for cancer
CN105916876A (en) * 2013-09-16 2016-08-31 分子医学研究中心责任有限公司 Mutant calreticulin for the diagnosis of myeloid malignancies
CN107148481A (en) * 2014-07-14 2017-09-08 苏黎世大学医学与自然科学院 The patient for suffering from BRAF positive cancers for determination is BRAF inhibitor non-response persons and is the tool and method of MAPK/ERK inhibitor respondents
KR20180029776A (en) * 2016-09-13 2018-03-21 경희대학교 산학협력단 A instrument on pattern identification for lung cancer using gene sequencing
JP2018143178A (en) * 2017-03-06 2018-09-20 国立大学法人三重大学 Genetic risk detection method for cardiovascular disease
CN107944224A (en) * 2017-12-06 2018-04-20 懿奈(上海)生物科技有限公司 Build method and the application of skin-related gene standard type database
CN110607371A (en) * 2019-10-14 2019-12-24 普瑞基准科技(北京)有限公司 Stomach cancer marker and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JIANWEI LIU等: ""Comprehensive investigation of the clinical significance and molecular mechanisms of plasmacytoma variant translocation 1 in sarcoma using genome-wide RNA sequencing data "", 《JOURNAL OF CANCER》 *
NULL: ""Gene: C10orf71 ENSG00000177354"", 《ENSEMBLE数据库》 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110607371A (en) * 2019-10-14 2019-12-24 普瑞基准科技(北京)有限公司 Stomach cancer marker and application thereof
CN110607371B (en) * 2019-10-14 2022-05-06 普瑞基准科技(北京)有限公司 Stomach cancer marker and application thereof
CN114525344A (en) * 2022-04-22 2022-05-24 普瑞基准科技(北京)有限公司 Kit for detecting or assisting in detecting tumor-related gene variation and application thereof
CN114561472A (en) * 2022-04-27 2022-05-31 普瑞基准科技(北京)有限公司 Kit for detecting or assisting in detecting tumor-related gene variation and application thereof

Also Published As

Publication number Publication date
CN112795647B (en) 2022-08-16

Similar Documents

Publication Publication Date Title
Yoo et al. Integrative analysis of genomic and transcriptomic characteristics associated with progression of aggressive thyroid cancer
Yan et al. Multi-region sequencing unveils novel actionable targets and spatial heterogeneity in esophageal squamous cell carcinoma
Bi et al. Genomic landscape of high-grade meningiomas
Fantini et al. A Carcinogen-induced mouse model recapitulates the molecular alterations of human muscle invasive bladder cancer
Chang et al. Genomic analysis of oesophageal squamous-cell carcinoma identifies alcohol drinking-related mutation signature and genomic alterations
Roy et al. Recurrent internal tandem duplications of BCOR in clear cell sarcoma of the kidney
CN112795647B (en) Tumor marker and application thereof
Kim et al. Mutational analysis of FOXL2 codon 134 in granulosa cell tumour of ovary and other human cancers
Waddell et al. Whole genomes redefine the mutational landscape of pancreatic cancer
Holbrook et al. Deep sequencing of gastric carcinoma reveals somatic mutations relevant to personalized medicine
Snuderl et al. Recurrent homozygous deletion of DROSHA and microduplication of PDE4DIP in pineoblastoma
Liang et al. Genome-wide characterization of pancreatic adenocarcinoma patients using next generation sequencing
Aihara et al. Genetic and epigenetic stability of oligodendrogliomas at recurrence
Nomura et al. Distinct molecular profile of diffuse cerebellar gliomas
Sakai et al. Genetic and epigenetic aberrations occurring in colorectal tumors associated with serrated pathway
Li et al. Germline and somatic mutations of multi-gene panel in Chinese patients with epithelial ovarian cancer: a prospective cohort study
Smith et al. Genetic and epigenetic profiling of CLL disease progression reveals limited somatic evolution and suggests a relationship to memory-cell development
Casey et al. Next generation sequencing and a new era of medicine
Choi et al. RTK-RAS pathway mutation is enriched in myeloid sarcoma
Chang et al. Whole exome sequencing of pediatric gastric adenocarcinoma reveals an atypical presentation of Li‐Fraumeni syndrome
Hájková et al. Synchronous endometrioid endometrial and ovarian carcinomas are biologically related: A clinico‑pathological and molecular (next generation sequencing) study of 22 cases
Zhu et al. The genomic and epigenomic evolutionary history of papillary renal cell carcinomas
Pira et al. Landscape of transcriptome variations uncovering known and novel driver events in colorectal carcinoma
Arriba et al. Intermediate-onset colorectal cancer: a clinical and familial boundary between both early and late-onset colorectal cancer
Berrino et al. Unique patterns of heterogeneous mismatch repair protein expression in colorectal cancer unveil different degrees of tumor mutational burden and distinct tumor microenvironment features

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
CB03 Change of inventor or designer information

Inventor after: Ji Jiafu

Inventor after: Li Ziyu

Inventor after: Gao Xiangyu

Inventor after: Ji Xuwo

Inventor after: Peng Xinxin

Inventor after: Li Zhe

Inventor after: Wei Bin

Inventor after: Wei Baoye

Inventor after: Dong Yu

Inventor before: Ji Jiafu

Inventor before: Dong Yu

Inventor before: Li Ziyu

Inventor before: Gao Xiangyu

Inventor before: Ji Xuwo

Inventor before: Liang Han

Inventor before: Peng Xinxin

Inventor before: Li Zhe

Inventor before: Wei Bin

Inventor before: Wei Baoye

CB03 Change of inventor or designer information
GR01 Patent grant
GR01 Patent grant