WO2017110764A1 - Test method for predicting effectiveness and safety of multi-kinase inhibitor, test kit, and biomarker - Google Patents

Test method for predicting effectiveness and safety of multi-kinase inhibitor, test kit, and biomarker Download PDF

Info

Publication number
WO2017110764A1
WO2017110764A1 PCT/JP2016/087859 JP2016087859W WO2017110764A1 WO 2017110764 A1 WO2017110764 A1 WO 2017110764A1 JP 2016087859 W JP2016087859 W JP 2016087859W WO 2017110764 A1 WO2017110764 A1 WO 2017110764A1
Authority
WO
WIPO (PCT)
Prior art keywords
treatment
concentration
vegf
ccl
regorafenib
Prior art date
Application number
PCT/JP2016/087859
Other languages
French (fr)
Japanese (ja)
Inventor
光邦 末永
信之 水沼
哲夫 馬島
啓之 清宮
Original Assignee
公益財団法人がん研究会
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 公益財団法人がん研究会 filed Critical 公益財団法人がん研究会
Priority to JP2017558133A priority Critical patent/JP6853789B2/en
Publication of WO2017110764A1 publication Critical patent/WO2017110764A1/en

Links

Images

Classifications

    • 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

Definitions

  • the present invention relates to a method for predicting the effectiveness and safety of a multikinase inhibitor targeting tumor cells, and a test kit.
  • the present invention particularly relates to the effectiveness of regorafenib, a method for predicting the risk of side effects, biomarkers used therefor, and kits.
  • Chemotherapy using an anticancer drug is a treatment method that is widely used together with surgery and radiation therapy as a tumor treatment method.
  • Chemotherapy using anticancer drugs involves treating cancer cells that may spread throughout the body or cancer cells that have the potential to metastasize by administering anticancer drugs orally or by injection. It is aimed.
  • Anticancer drugs include platinum agents, alkylating agents, plant alkaloids that act on mitotic cells, and specific properties of cancer cells at the molecular level. There are various types such as targeted drugs.
  • side effects often occur because any anticancer agent acts on normal cells. Symptoms that often appear as side effects include decreased white blood cells and platelets, vomiting / nausea, hair loss, general malaise, and stomatitis. Since side effects occur with most anticancer agents, anticancer agents with reduced side effects and pharmaceuticals that reduce side effects have been developed. As a result, side effects have been reduced compared to before, but the side effects caused by the administration of anticancer agents are a major problem in chemotherapy.
  • Patent Documents 1 and 2 disclose a method for predicting the risk of side effects of docetaxel used for treatment of refractory breast cancer, non-small cell lung cancer, ovarian cancer, etc. by detecting single nucleotide polymorphisms present on SLCO1B3 gene and ABCC2 gene. Is described.
  • Patent Document 2 describes a method for predicting side effects by analyzing the correlation between side effects caused by fluorouracil and cisplatin used in a wide variety of treatments and cytokine gene polymorphisms.
  • Non-patent document 1 describes that the therapeutic effect of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) administration on non-small cell lung cancer patients is correlated with serum HGF concentration. It is disclosed.
  • Non-Patent Document 2 FOLFIRI therapy for patients with colorectal cancer (Fol nic acid, F luorouracil, Iri notecan) performs a therapy plus bevacizumab, analyzes the correlation between efficacy and biomarker for therapeutic It is described. In particular, it has been disclosed that changes in serum VEGF-A concentration correlate with therapeutic efficacy.
  • EGFR-TKI epidermal growth factor receptor tyrosine kinase inhibitor
  • Patent Documents 1 and 2 require that DNA is extracted from serum and PCR is performed, so that protein in serum is directly measured by immunoassay represented by ELISA. Compared to the method, there are many processes and complicated.
  • Regorafenib is a multikinase inhibitor that inhibits multiple protein kinase activities.
  • Legorafenib is an angiogenesis-related kinase (VEGFR1-3, TIE2), a tumor microenvironment-related stromal kinase (PDGFR ⁇ , FGFR), a tumorigenesis-related kinase (KIT, RET, RAF-1, BRAF) Have been shown to inhibit multiple protein kinase activities.
  • VEGFR1-3 angiogenesis-related kinase
  • PDGFR ⁇ tumor microenvironment-related stromal kinase
  • KIT tumorigenesis-related kinase
  • RET RAF-1
  • BRAF tumorigenesis-related kinase
  • Regorafenib is a side effect (including abnormal laboratory values) in 465 cases (93.0%) of 500 cases (including 65 Japanese) in a global phase III clinical trial for patients with colorectal cancer Was recognized.
  • the number of cases of incidence of major side effects (incidence rate) was 225 cases of hand-foot syndrome (45.0%), 169 cases of diarrhea (33.8%), 152 cases of decreased appetite (30.4%), and 145 cases of fatigue (29 0.0%), dysphonia 142 cases (28.4%), hypertension 139 cases (27.8%), and rash 113 cases (22.6%).
  • limb syndrome toxic epidermal necrolysis (TEN), mucocutaneous eye syndrome (Stevens-Johnson syndrome), erythema multiforme, fulminant hepatitis, liver failure, liver dysfunction, Jaundice, bleeding (gastrointestinal bleeding, hemoptysis, pulmonary bleeding, intraperitoneal bleeding, vaginal bleeding, cerebral bleeding, nasal bleeding, hematuria, etc.), interstitial lung disease, thromboembolism (myocardial ischemia, myocardial infarction, etc.), hypertension, hypertension Crisis, post-plastic leukoencephalopathy, gastrointestinal perforation, gastrointestinal fistula, thrombocytopenia may occur.
  • the object of the present invention is to extract a marker for predicting the effect of regorafenib and a marker relating to the onset of an adverse event, and to use it for actual clinical treatment. It is another object of the present invention to provide an inspection method and an inspection kit using the marker.
  • the present invention relates to the following test method, test kit, and biomarker related to the effect prediction of regorafenib.
  • a test method for predicting the effect of administration of Regorafenib comprising measuring CCL-5 concentration in a sample collected from a patient.
  • the test method according to (1) wherein the sample is blood, plasma, serum, urine, ascites, or pleural effusion.
  • the test method according to (3) wherein when the CCL-5 concentration is lower than a predetermined value, it is determined that improvement in progression-free survival (PFS) and overall survival (OS) can be expected Method.
  • PFS progression-free survival
  • OS overall survival
  • a VEGF-A concentration in a patient sample is measured before the start of treatment and early after the start of the first treatment.
  • test method according to any one of (1) to (6), wherein at least one of Ang-2, bFGF, and CCL-2 in a patient sample before the start of treatment is measured, A test method to determine the risk of adverse events.
  • a test kit for predicting the effect of regorafenib administration comprising a reagent for measuring CCL-5 concentration in a patient sample.
  • the reagent is an immunoassay reagent.
  • a biomarker for predicting the effect of regorafenib administration which is CCL-5 and / or VEGF-A.
  • a biomarker for predicting an adverse event caused by administration of regorafenib wherein the biomarker comprises at least one of CCL-5, VEGF-A, Ang-2, bFGF, and CCL-2.
  • the present invention provides a marker for predicting the effects of regorafenib and a marker for the onset of adverse events, which can be used for actual clinical treatment.
  • a marker for predicting the effects of regorafenib and a marker for the onset of adverse events which can be used for actual clinical treatment.
  • serum CCL-5 Chemokine (CC motif) ligand-5)
  • VEGF-A vascular endothelial growth factor-A, vascular endothelial growth factor-A
  • the effect can be predicted more accurately at an early stage of treatment. be able to.
  • FIG. 3 (A) shows the ROC curve of tumor shrinkage and CCL-5 value.
  • FIG. 3 (B) shows CCL-5 ⁇ 59.96 (ng / mL), CCL-5> 59.96 (ng / mL).
  • the progression-free survival period is shown in FIG. 3 (C).
  • FIG. 4 (A) shows the relationship between the decrease on day 21 after the start of VEGF-A treatment and progression-free survival
  • FIG. 4 (B) shows the decrease on day 21 after the start of treatment with VEGF-A and the primary disease. The relationship between the increase in exacerbation and progression-free survival is shown.
  • FIG. 5 (A) shows that the CCL-5 concentration before the start of treatment is less than or equal to the cut-off value, the VEGF-A concentration after the start of treatment decreases, and the CCL-5 concentration before the start of treatment is higher than the cut-off value.
  • concentration after a treatment start increases is shown.
  • FIG. 5B further shows a group not classified into both. The figure which analyzed the total survival time combining the CCL-5 density
  • FIG. 6A shows that the CCL-5 concentration before the start of treatment is less than or equal to the cut-off value, the VEGF-A concentration after the start of treatment decreases, and the CCL-5 concentration before the start of treatment is higher than the cut-off value.
  • concentration after a treatment start increases is shown.
  • FIG. 6B further shows a group that is not classified into both. The figure which analyzed the progression-free survival time combining CCL-5 density
  • FIG. 7A shows a group in which the CCL-5 concentration before the start of treatment is below the cut-off value, the VEGF-A concentration after the start of treatment decreases, and the VEGF-A concentration in the exacerbation stage increases, and the CCL before the start of treatment.
  • the comparison is made with the group in which the -5 concentration is higher than the cut-off value, the VEGF-A concentration is increased after the start of the treatment, and the VEGF-A concentration is decreased in the exacerbation stage.
  • FIG. 7B further shows a group that is not classified into both.
  • FIG. 8A shows a group in which the CCL-5 concentration before the start of treatment is below the cut-off value, the VEGF-A concentration after the start of treatment decreases, the VEGF-A concentration in the exacerbation stage increases, and the CCL before the start of treatment.
  • the comparison is made with the group in which the -5 concentration is higher than the cut-off value, the VEGF-A concentration is increased after the start of the treatment, and the VEGF-A concentration is decreased in the exacerbation stage.
  • FIG. 8B further shows a group not classified into both.
  • an adverse event refers to any undesirable or unintended sign, symptom, or illness that has occurred in a patient to whom a drug has been administered, including those in which the causal relationship with the drug is not clear. It is not limited to side effects with known causal relationships.
  • progression-free survival Progression Free; Survival, PFS
  • overall survival Overall Survival; OS
  • the progression-free survival period is from the date of start of administration to the date on which the exacerbation (PD) is determined or the death date due to any cause (regardless of cause of death), whichever comes first.
  • Surviving cases that are not judged to be exacerbated are treated as censored on the last day when it is confirmed that there is no exacerbation. In other words, if an exacerbation is not confirmed when the event is canceled due to an adverse event, it is treated as being terminated.
  • “Overall survival period (OS)” means the total survival period from the start date of administration to the date of death due to any cause. In surviving cases, the date of confirmation of final survival is censored, and in the case of inability to follow up, the censored date is determined to be the last date of confirmation of survival before becoming untraceable.
  • the sample refers to blood, plasma, serum, urine, ascites, pleural effusion. This is because it is already known that the marker of the present invention is contained in these samples (Non-Patent Documents 4 to 8).
  • blood, plasma, and serum are preferable as samples because the biomarker of the present invention can be measured with high sensitivity.
  • the sample before starting treatment refers to a sample before starting treatment with regorafenib for patients who have become refractory or intolerant to standard chemotherapy.
  • the sample of the 21st day is used for the sample at the early stage after the start of the first treatment in the Examples, the first cycle is one cycle (one cycle is provided with a one week rest period after oral administration for 3 weeks every day). Any sample may be used as long as it is a sample. Considering the effect of drug administration, the samples on the 14th to 28th days are preferable.
  • any measurement method may be used as long as the protein amount of the biomarker in the sample can be measured. Measurement is preferably performed by immunoassay because measurement sensitivity is high and testing can be performed relatively easily.
  • immunoassay for example, radioimmunoassay (RIA), fluorescence immunoassay (FIA), fluorescence polarization immunoassay (FPIA), chemiluminescence immunoassay (CLIA), and the like can be used.
  • the “cut-off value” is a value that can satisfy both high diagnostic sensitivity (adverse event rate) and high diagnostic specificity when a disease is determined based on the value. For example, a value showing a high positive rate in an individual who has developed an adverse event and a high negative rate in an individual who has not developed an adverse event can be set as the cutoff value. Since the specific cut-off value varies depending on the sample and assay system used, it can be appropriately determined according to each assay system.
  • the cut-off value calculation method may be determined by a method known in this field (for example, see Non-Patent Documents 9 to 11).
  • the kit of the present invention can contain an antibody for detecting CCL-5 and VEGF-A, which are biomarkers found in the present invention, a reagent for detecting this, and the like.
  • the detection reagent includes a secondary antibody, a substrate agent, and a labeling substance (for example, fluorescent dye, enzyme). Moreover, these elements can also be mixed beforehand as needed. Further, it may include a solid phase such as a microtiter plate, a reaction container, a buffer for diluting a washing solution or an antibody, a positive control, a negative control, an instruction describing a protocol, and the like.
  • Regorafenib is a multikinase inhibitor, as described above, and has been suggested to act on cancer cell environmental signals related to signal transduction and angiogenesis. However, it was not clear which of these factors was directly affected by regorafenib treatment. Therefore, the present inventors analyzed changes in factors secreted from cancer cells detected by microarray gene expression analysis or ELISA method, in addition to candidates selected from factors involved in angiogenesis and inflammatory cytokines. .
  • HCT-15, HCT-116 and HT-29 available from ATCC were used as human colon cancer cell lines.
  • the above colon cancer cell lines were seeded at a cell density of 9000 cells / mL using 10% FBS-RPMI medium, 24 hours later, regorafenib was added to a concentration of 0 ⁇ M, 1 ⁇ M, and 3 ⁇ M and cultured for 72 hours. Cell proliferation was analyzed by MTS assay (Promega). The results are shown in FIG.
  • cell growth is suppressed in a concentration-dependent manner by adding regorafenib.
  • the HT-29 and HCT-116 cell lines are extremely strong and suppress cell growth and are highly sensitive to regorafenib.
  • the HCT-15 cell line is a cell line that is less resistant to cell growth than other cell lines and has resistance to regorafenib. It became clear.
  • VEGF-A was found as a factor having a significant change in the secretion amount in correlation with the regorafenib sensitivity of the cells (FIG. 2).
  • Regorafenib was added to colorectal cancer cell lines HCT-15, HCT-116, and HT-29, and VEGF-A concentration secreted into the culture medium was measured 48 hours later by ELISA (Quantikine ELISA kit from R & D Systems) ).
  • the VEGF-A concentration is shown with the VEGF-A concentration in the culture medium being 100% when cultured without adding regorafenib in each cultured cell line.
  • VEGF-A in colon cancer cells Information on the expression of VEGF-A in colon cancer cells can be obtained from Oncomine database (https://www.oncomine.org/resource/login.html). According to this, expression of VEGF-A is increased in colon cancer cells.
  • Kumar et al. Non-patent Document 12
  • the concentration of VEGF-A is significantly higher in the serum of colorectal cancer patients than in the healthy human serum, and the VEGF-A level is higher in advanced cancer.
  • VEGF-A In addition to VEGF-A, CCL-2, CCL-5, IL-8 (interleukin-8), PlGF (Placent Growth Factor), Ang-1 (angiopoietin-1), Ang-2 (angiopoietin-2), bFGF ( Analysis was performed using patient serum using basic fibroblast growth factor), SDF-1 (stromal cell-derived factor 1), and PDGF ⁇ (platelet-derived growth factor beta) as candidate factors.
  • IL-8 interleukin-8
  • PlGF Placent Growth Factor
  • Ang-1 angiopoietin-1
  • Ang-2 angiopoietin-2
  • bFGF Analysis was performed using patient serum using basic fibroblast growth factor
  • SDF-1 stromal cell-derived factor 1
  • PDGF ⁇ platelet-derived growth factor beta
  • the amount of cytokines such as CCL-5 and VEGF-A in the serum was determined according to the attached protocol by the sandwich ELISA method using Quantikine ELISA kit of R & D Systems.
  • the buffer attached to the kit was added to a 96-well plate coated with an antibody against each cytokine, and then a serum sample and a cytokine preparation for preparing a calibration curve (dilution series) were added and reacted at room temperature for 2 hours. . Thereafter, after washing three times with a washing buffer, an HRP-labeled antibody against each cytokine was added, and further reacted at room temperature for 1-2 hours. Thereafter, after washing three times with a washing buffer, a coloring solution was added, and after reacting at room temperature for 20-25 minutes, a reaction stop solution was added, and OD450 and OD570 (background) were measured using a plate reader. . Each sample was tested in triplicate, and the cytokine concentration was calculated based on the calibration curve for the value obtained by subtracting the background.
  • ELISA was conducted at 3 points before the start of the first treatment of regorafenib, 21 days after the start of the first treatment, and at the time of the decision to stop the treatment, and whether each candidate factor was correlated with the occurrence of an adverse event was analyzed. .
  • CCL-5 and VEGF-A were correlated with efficacy and adverse events.
  • CCL-5 and VEGF-A levels in the sample are related to efficacy and incidence of adverse events that are particularly common in Japanese, ie hand-foot syndrome, hypertension, liver dysfunction, hyperbilirubinemia, thrombocytopenia It became clear that it was an effective predictive marker. The results will be described in detail below.
  • FIG. 3 (A) shows an ROC curve created for tumor shrinkage and serum CCL-5 concentration. From this result, if the CCL-5 concentration in the serum before the start of treatment is CCL-5 ⁇ 59.96 (ng / mL), a tumor reducing effect can be expected by administration of regorafenib.
  • the VEGF-A concentration decreased on the 21st day, and the pattern of increase during exacerbation of the original disease improved the progression-free survival period, and the average value of the progression-free survival period was 146 days.
  • FIG. 4 (B) There was a statistically significant correlation between the VEGF-A concentration on day 21 after the start of treatment and the concentration at the time of exacerbation of the original disease.
  • it was not statistically significant it was recognized that the same tendency was shown also in the whole survival period. That is, it became clear that the change in serum VEGF-A level after the start of treatment is a factor predicting the therapeutic effect.
  • the patient group which is not classified into the above was analyzed as a Medium group. From the Kaplan-Meier curve, the progression-free survival period of the Medium group was a result intermediate between the Good group and the Poor group (FIG. 5B). Mean values of progression-free survival in the Good group, Medium group, and Poor group were 188 days, 69 days, and 70 days, respectively.
  • serum CCL-5 and VEGF-A can be used alone or in combination as a marker for predicting the effect of regorafenib monotherapy before treatment or at the beginning of treatment.
  • AE indicates an adverse event (Adverse Event)
  • T-Bil indicates the total amount of bilirubin.
  • AST is an abbreviation for aspartate aminotransferase and ALT is an abbreviation for alanine aminotransferase, both of which are test values serving as indicators of liver function.
  • HT represents hypertension
  • HFS represents hand-foot syndrome.
  • Ang-2, bFGF, and CCL-2 are as described above.
  • Ang-2, bFGF, and CCL-2 concentrations before the start of treatment can be predictive markers for the onset of adverse events involved in treatment performance.
  • AST values of grade 2 ⁇ were particularly correlated when CCL-5 concentration decreased after treatment, and liver function abnormalities were frequently developed.
  • Combination of CCL-5 concentration (below CO value) before treatment start and change in VEGF-A concentration after treatment start is associated with the development of hyperbilirubinemia (grade2 ⁇ ) and thrombocytopenia (grade1 ⁇ , grade2 ⁇ ) It became clear that Similar results were obtained with VEGF alone.
  • CCL-5 and VEGF-A can be used as predictive markers for the effect of pretreatment or early treatment onset of adverse events with high incidence in Japanese in regorafenib monotherapy in salvage therapy.
  • the correlation between these adverse events and CCL-5 and VEGF-A is not inconsistent with the correlation between progression-free survival and overall survival.
  • Treatment can be performed.
  • an adverse event can be predicted by measuring the Ang-2, bFGF, and CCL-2 concentrations before starting treatment.
  • regorafenib was analyzed in detail, but application to other multikinase inhibitors such as sorafenib and sunitinib which are multikinase inhibitors having similar structures and activities of the compounds can also be expected.
  • anticancer drug treatment is performed only on patients who are effective in treatment by selecting patients who respond well to anticancer drug administration and chemotherapy. The risk of serious side effects can be avoided.

Abstract

Provided are a test method for predicting the effect of administering Regorafenib, and a test kit. Measurements are taken of the CCL-5 concentration and the VEGF-A concentration in a specimen collected from a patient, whereby the effectiveness of treatment on the patient and the risk of adverse events are estimated.

Description

マルチキナーゼ阻害剤の有効性と安全性を予測する検査方法、検査キット、及びバイオマーカーTest method, test kit, and biomarker for predicting efficacy and safety of multikinase inhibitor
 本発明は腫瘍細胞を標的とするマルチキナーゼ阻害剤の有効性と安全性を予測する方法、及び検査キットに関する。マルチキナーゼ阻害剤の中でも、特にレゴラフェニブ(Regorafenib)の有効性、副作用のリスクを予測する方法、これに用いるバイオマーカー、及びキットに関する。 The present invention relates to a method for predicting the effectiveness and safety of a multikinase inhibitor targeting tumor cells, and a test kit. Among multikinase inhibitors, the present invention particularly relates to the effectiveness of regorafenib, a method for predicting the risk of side effects, biomarkers used therefor, and kits.
 抗がん剤を用いる化学療法は、腫瘍の治療方法として、手術、放射線療法とともに広く行われている治療方法である。抗がん剤を用いて行う化学療法は、抗がん剤を経口、又は注射により投与することにより全身に広がる可能性のある癌細胞や、転移の可能性のある癌細胞を治療することを目的としている。 Chemotherapy using an anticancer drug is a treatment method that is widely used together with surgery and radiation therapy as a tumor treatment method. Chemotherapy using anticancer drugs involves treating cancer cells that may spread throughout the body or cancer cells that have the potential to metastasize by administering anticancer drugs orally or by injection. It is aimed.
 抗がん剤には、分裂時の細胞に作用するプラチナ剤、アルキル化剤、植物アルカロイド、癌細胞の持つ特異的な性質を分子レベルでとらえ、それを標的として癌細胞に効率良く作用する分子標的薬等様々な種類がある。しかしながら、いずれの抗がん剤も正常細胞に対して少なからず作用することから副作用が生じることが多い。副作用としてよく表れる症状には、白血球や血小板の減少、嘔吐・悪心、脱毛、全身倦怠感、口内炎が知られている。ほとんどの抗がん剤で副作用が生じることから、副作用が軽減された抗がん剤、副作用を軽減する医薬が開発されてきている。その結果、副作用は以前に比べて軽減されてきているものの、抗がん剤投与によって生じる副作用は化学療法において大きな問題となっている。 Anticancer drugs include platinum agents, alkylating agents, plant alkaloids that act on mitotic cells, and specific properties of cancer cells at the molecular level. There are various types such as targeted drugs. However, side effects often occur because any anticancer agent acts on normal cells. Symptoms that often appear as side effects include decreased white blood cells and platelets, vomiting / nausea, hair loss, general malaise, and stomatitis. Since side effects occur with most anticancer agents, anticancer agents with reduced side effects and pharmaceuticals that reduce side effects have been developed. As a result, side effects have been reduced compared to before, but the side effects caused by the administration of anticancer agents are a major problem in chemotherapy.
 また、副作用の種類や重篤度は、使用する抗がん剤によって異なるだけではなく、個人差が大きいことが知られている。さらに、副作用だけではなく、投与した薬物と因果関係がはっきりしない有害事象が生じることも知られている。さらに抗がん剤の効果についても個人差があるため、同じ種類の腫瘍であっても、患者によってはあまり効果を得られず、副作用が強く生じる場合もある。したがって、抗がん剤投与によって顕著な効果が期待できるか、また、重篤な副作用や有害事象が生じるかを、抗がん剤を投与する前に判断することができれば、より効果が高く、副作用、有害事象の少ない抗がん剤を選択することができる。薬剤投与による有効性と安全性を予測することができれば、患者にとって大きなメリットとなる。 Also, it is known that the types and severity of side effects are not only different depending on the anticancer agent used, but also vary greatly from individual to individual. Furthermore, it is known that not only side effects but also adverse events whose causal relationship is not clear. Furthermore, because there are individual differences in the effects of anticancer agents, even for the same type of tumor, some patients may not be very effective and may have strong side effects. Therefore, if the anticancer drug administration can be expected to have a significant effect and whether serious side effects or adverse events occur can be determined before administering the anticancer drug, Anticancer drugs with few side effects and adverse events can be selected. If the effectiveness and safety of drug administration can be predicted, it will be a great advantage for patients.
 近年、抗がん剤治療により変化する血液中のマーカーと治療の有効性との関係について解析が行われるようになった(特許文献1、2、非特許文献1、2)。特許文献1には、難治性乳癌、非小細胞肺癌、卵巣癌などの治療に使用されるドセタキセルの副作用の危険性をSLCO1B3遺伝子及びABCC2遺伝子上に存在する一塩基多型の検出により予測する方法が記載されている。特許文献2には、広範な種類の治療に使用されているフルオロウラシル、シスプラチンによる副作用とサイトカイン遺伝子多型の相関を解析し、副作用の予測を行う方法が記載されている。 In recent years, analysis has been conducted on the relationship between markers in blood that change due to anticancer drug treatment and the effectiveness of treatment (Patent Documents 1 and 2, Non-Patent Documents 1 and 2). Patent Document 1 discloses a method for predicting the risk of side effects of docetaxel used for treatment of refractory breast cancer, non-small cell lung cancer, ovarian cancer, etc. by detecting single nucleotide polymorphisms present on SLCO1B3 gene and ABCC2 gene. Is described. Patent Document 2 describes a method for predicting side effects by analyzing the correlation between side effects caused by fluorouracil and cisplatin used in a wide variety of treatments and cytokine gene polymorphisms.
 非特許文献1には、非小細胞肺癌患者に対する上皮成長因子受容体チロシンキナーゼ阻害薬(Epidermal Growth Factor Receptor‐Tyrosine Kinase Inhibitor:EGFR‐TKI)投与の治療効果が血清HGF濃度と相関があることが開示されている。非特許文献2には、結腸直腸癌の患者に対してFOLFIRI療法(Folnic acid、luorouracil、Irinotecan)にベバシズマブを加えた治療法を行い、治療に対する有効性とバイオマーカーとの相関を解析したことが記載されている。特に、血清中のVEGF‐A濃度の変化が治療の有効性と相関することが開示されている。 Non-patent document 1 describes that the therapeutic effect of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) administration on non-small cell lung cancer patients is correlated with serum HGF concentration. It is disclosed. Non-Patent Document 2, FOLFIRI therapy for patients with colorectal cancer (Fol nic acid, F luorouracil, Iri notecan) performs a therapy plus bevacizumab, analyzes the correlation between efficacy and biomarker for therapeutic It is described. In particular, it has been disclosed that changes in serum VEGF-A concentration correlate with therapeutic efficacy.
 しかし、特許文献1及び2に開示されている遺伝子多型による解析は、血清からDNAを抽出し、PCRを行う必要があることから、ELISAに代表される免疫測定により血清中のタンパク質を直接測定する方法に比べ過程が多く煩雑である。 However, the analysis by genetic polymorphism disclosed in Patent Documents 1 and 2 requires that DNA is extracted from serum and PCR is performed, so that protein in serum is directly measured by immunoassay represented by ELISA. Compared to the method, there are many processes and complicated.
 また、上記先行技術文献に開示されているように、特定の抗がん剤や化学療法レジメンに関しては、抗がん剤投与による副作用のリスクが解析されている。しかしながら、多くの抗がん剤は治療前に有効性や副作用を検討するマーカーがないため、化学療法を行いながら、治療の有効性と副作用の強さを検討しているのが実情である。 Also, as disclosed in the above prior art documents, the risk of side effects caused by administration of anticancer agents has been analyzed for specific anticancer agents and chemotherapy regimens. However, since many anticancer agents do not have markers for examining the efficacy and side effects before treatment, it is the actual situation that the efficacy of treatment and the strength of side effects are being examined while performing chemotherapy.
 レゴラフェニブは、複数のプロテインキナーゼ活性を阻害するマルチキナーゼ阻害薬である。レゴラフェニブは、血管新生に関連するキナーゼ(VEGFR1‐3、TIE2)、腫瘍微小環境に関連する間質のキナーゼ(PDGFRβ、FGFR)、腫瘍形成に関連するキナーゼ(KIT、RET、RAF‐1、BRAF)など、複数のプロテインキナーゼ活性を阻害することが明らかにされている。 Regorafenib is a multikinase inhibitor that inhibits multiple protein kinase activities. Legorafenib is an angiogenesis-related kinase (VEGFR1-3, TIE2), a tumor microenvironment-related stromal kinase (PDGFRβ, FGFR), a tumorigenesis-related kinase (KIT, RET, RAF-1, BRAF) Have been shown to inhibit multiple protein kinase activities.
 本邦では2013年3月25日に治癒切除不能な進行・再発の結腸・直腸癌の効能・効果に対して承認された。実臨床では治癒切除不能な進行・再発の結腸・直腸癌に対して既存の標準化学療法に対して病勢進行が認められた場合に最終ラインとして単剤療法で用いられることが一般的である。また、2013年8月20日、化学療法後に増悪した消化管間質腫瘍(Gastrointestinal Stromal Tumor:GIST)に対しても承認されている。 In Japan, it was approved on March 25, 2013 for the efficacy and effects of unresectable advanced / recurrent colorectal cancer. In clinical practice, it is common to use single-agent therapy as the last line when progression of relapsed colorectal cancer that cannot be curatively resected has progressed against existing standard chemotherapy. On August 20, 2013, it was also approved for gastrointestinal stromal tumor (GIST) that worsened after chemotherapy.
 レゴラフェニブは、結腸・直腸癌患者を対象とした国際共同第III相臨床試験において、500例中(日本人65例を含む)465例(93.0%)に副作用(臨床検査値異常を含む)が認められた。主な副作用の発現例数(発現率)は,手足症候群225例(45.0%)、下痢169例(33.8%)、食欲減退152例(30.4%)、疲労145例(29.0%)、発声障害142例(28.4%)、高血圧139例(27.8%)、発疹113例(22.6%)であった。 Regorafenib is a side effect (including abnormal laboratory values) in 465 cases (93.0%) of 500 cases (including 65 Japanese) in a global phase III clinical trial for patients with colorectal cancer Was recognized. The number of cases of incidence of major side effects (incidence rate) was 225 cases of hand-foot syndrome (45.0%), 169 cases of diarrhea (33.8%), 152 cases of decreased appetite (30.4%), and 145 cases of fatigue (29 0.0%), dysphonia 142 cases (28.4%), hypertension 139 cases (27.8%), and rash 113 cases (22.6%).
 また、消化管間質腫瘍患者を対象とした国際共同第III相臨床試験において、132例中(日本人12例を含む)130例(98.5%)に副作用(臨床検査値異常を含む)が認められた。主な副作用の発現例数(発現率)は、手足症候群87例(65.9%)、高血圧64例(48.5%)、下痢53例(40.2%)、発声障害44例(33.3%)、疲労39例(29.5%)、発疹38例(28.8%)食欲減退28例(21.2%)であった。 In an international joint phase III clinical trial for patients with gastrointestinal stromal tumors, 130 (98.5%) of 132 cases (including 12 Japanese) had side effects (including abnormal laboratory values). Was recognized. The number of cases of incidence of major side effects (incidence rate) was 87 cases (65.9%) for hand-foot syndrome, 64 cases (48.5%) for hypertension, 53 cases (40.2%) for diarrhea, 44 cases for dysphonia (33) 3%), 39 cases (29.5%) of fatigue, 38 cases (28.8%) of rash, and 28 cases (21.2%) of decreased appetite.
 また、重大な副作用として、手足症候群、中毒性表皮壊死融解症(Toxic Epidermal Necrolysis:TEN)、皮膚粘膜眼症候群(Stevens‐Johnson症候群)、多形紅斑、劇症肝炎、肝不全、肝機能障害、黄疸、出血(消化管出血、喀血、肺出血、腹腔内出血、膣出血、脳出血、鼻出血、血尿等)、間質性肺疾患、血栓塞栓症(心筋虚血、心筋梗塞等)、高血圧、高血圧クリーゼ、可塑性後白質脳症、消化管穿孔、消化管瘻、血小板減少が現れることがある。 In addition, as serious side effects, limb syndrome, toxic epidermal necrolysis (TEN), mucocutaneous eye syndrome (Stevens-Johnson syndrome), erythema multiforme, fulminant hepatitis, liver failure, liver dysfunction, Jaundice, bleeding (gastrointestinal bleeding, hemoptysis, pulmonary bleeding, intraperitoneal bleeding, vaginal bleeding, cerebral bleeding, nasal bleeding, hematuria, etc.), interstitial lung disease, thromboembolism (myocardial ischemia, myocardial infarction, etc.), hypertension, hypertension Crisis, post-plastic leukoencephalopathy, gastrointestinal perforation, gastrointestinal fistula, thrombocytopenia may occur.
 さらに、これら特有の副作用は、日本人とそれ以外での比較を行ったところ、日本人の発症率が高いことも明らかであった。重大な副作用の一つである肝機能障害は日本人での死亡報告もあり、適性使用ガイドでは治療中の厳重な経過観察が推奨されている。レゴラフェニブの有効性と副作用に関する予測マーカーの研究は国際共同第III相臨床試験でも行われたが、実臨床で有用な結果は得られていない(非特許文献3)。 Furthermore, when these side effects were compared between Japanese and others, it was clear that the incidence of Japanese was high. Liver dysfunction, which is one of the serious side effects, has been reported in Japanese deaths, and strict follow-up during treatment is recommended in the appropriate use guide. Although research on predictive markers for the efficacy and side effects of regorafenib was conducted in an international phase III clinical trial, no useful results were obtained in actual clinical practice (Non-patent Document 3).
特開2009‐240232号公報JP 2009-240232 A 特開2007‐006744号公報JP 2007-006744 A
 本発明は、レゴラフェニブの効果予測マーカーと有害事象の発症に関するマーカーを抽出し、実臨床での治療に役立てることを課題とする。また、当該マーカーを用いた検査方法、検査キットを提供することを課題とする。 The object of the present invention is to extract a marker for predicting the effect of regorafenib and a marker relating to the onset of an adverse event, and to use it for actual clinical treatment. It is another object of the present invention to provide an inspection method and an inspection kit using the marker.
 本発明は、レゴラフェニブの効果予測に関する以下の検査方法、検査キット、及びバイオマーカーに関する。 The present invention relates to the following test method, test kit, and biomarker related to the effect prediction of regorafenib.
(1)レゴラフェニブ(Regorafenib)投与による効果を予測するための検査方法であって、患者より採取された試料中のCCL‐5濃度を測定することを特徴とする検査方法。
(2)(1)記載の検査方法であって、前記試料が血液、血漿、血清、尿、腹水、胸水であることを特徴とする検査方法。
(3)(1)又は(2)記載の検査方法であって、前記試料が治療開始前のものであることを特徴とする検査方法。
(4)(3)記載の検査方法であって、CCL‐5濃度が所定値より低い場合には、無増悪生存期間(PFS)及び全生存期間(OS)の改善が期待できると判定する検査方法。
(5)(1)、(2)、又は(4)のいずれか1つに記載の検査方法であって、患者試料中の治療開始前と初回治療開始後早期のVEGF‐A濃度を測定し、治療開始後のVEGF‐A濃度が治療開始前と比較して減少傾向にある場合には、PFS及びOSの改善が期待できると判定する検査方法。
(6)(5)記載の検査方法であって、原病増悪時の患者試料中のVEGF‐A濃度を測定し、初回治療開始後早期のVEGF‐A濃度と比較して増加している場合には、PFS及びOSの改善が期待できると判定する検査方法。
(7)(1)~(6)のいずれか1つに記載の検査方法であって、治療開始前の患者試料中のAng‐2、bFGF、CCL‐2のうち少なくとも1つを測定し、有害事象が生じるリスクを判定する検査方法。
(8)レゴラフェニブ投与による効果を予測するための検査キットであって、患者試料中のCCL‐5濃度を測定するための試薬を含むことを特徴とする検査キット。
(9)(8)記載の検査キットであって、さらに、VEGF‐A濃度を測定するための試薬を含むことを特徴とする検査キット。
(10)(8)又は(9)記載の検査キットであって、Ang‐2、bFGF、CCL‐2のうち少なくとも1つの濃度を測定するための試薬を含むことを特徴とするキット。
(11)(8)~(10)いずれか1つ記載のキットであって、前記試薬がイムノアッセイ用の試薬であることを特徴とするキット。
(12)レゴラフェニブ投与による効果を予測するためのバイオマーカーであって、CCL‐5及び/又はVEGF‐Aであることを特徴とするバイオマーカー。
(13)レゴラフェニブ投与による有害事象を予測するためのバイオマーカーであって、CCL-5、VEGF‐A、Ang‐2、bFGF、CCL‐2の少なくとも1つからなることを特徴とするバイオマーカー。
(1) A test method for predicting the effect of administration of Regorafenib, wherein the test method comprises measuring CCL-5 concentration in a sample collected from a patient.
(2) The test method according to (1), wherein the sample is blood, plasma, serum, urine, ascites, or pleural effusion.
(3) The inspection method according to (1) or (2), wherein the sample is one before the start of treatment.
(4) The test method according to (3), wherein when the CCL-5 concentration is lower than a predetermined value, it is determined that improvement in progression-free survival (PFS) and overall survival (OS) can be expected Method.
(5) The test method according to any one of (1), (2), or (4), wherein a VEGF-A concentration in a patient sample is measured before the start of treatment and early after the start of the first treatment. A test method for determining that an improvement in PFS and OS can be expected when the VEGF-A concentration after the start of treatment tends to be lower than that before the start of treatment.
(6) The test method according to (5), wherein the VEGF-A concentration in the patient sample at the time of the exacerbation of the primary disease is measured and increased compared to the VEGF-A concentration early after the start of the first treatment Is an inspection method for determining that improvement of PFS and OS can be expected.
(7) The test method according to any one of (1) to (6), wherein at least one of Ang-2, bFGF, and CCL-2 in a patient sample before the start of treatment is measured, A test method to determine the risk of adverse events.
(8) A test kit for predicting the effect of regorafenib administration, comprising a reagent for measuring CCL-5 concentration in a patient sample.
(9) The test kit according to (8), further comprising a reagent for measuring a VEGF-A concentration.
(10) The test kit according to (8) or (9), comprising a reagent for measuring at least one concentration of Ang-2, bFGF, and CCL-2.
(11) The kit according to any one of (8) to (10), wherein the reagent is an immunoassay reagent.
(12) A biomarker for predicting the effect of regorafenib administration, which is CCL-5 and / or VEGF-A.
(13) A biomarker for predicting an adverse event caused by administration of regorafenib, wherein the biomarker comprises at least one of CCL-5, VEGF-A, Ang-2, bFGF, and CCL-2.
 本発明によりレゴラフェニブの効果予測マーカーと有害事象の発症に関するマーカーが得られたことで、実臨床での治療に役立てることができる。具体的には、予め血清中のCCL‐5(Chemokine(C‐C motif)ligand‐5)を測定することによって、治療に対して反応性が高く、重篤な有害事象、副作用のリスクが低い患者を選択して治療を行うことが可能となる。さらに、治療前、治療後21日目の血清中のVEGF-A(血管内皮増殖因子‐A、vascular endothelial growth factor‐A)の濃度を比較することによって、治療開始早期により正確な効果予測を行うことができる。 The present invention provides a marker for predicting the effects of regorafenib and a marker for the onset of adverse events, which can be used for actual clinical treatment. Specifically, by measuring serum CCL-5 (Chemokine (CC motif) ligand-5) in advance, it is highly responsive to treatment and has a low risk of serious adverse events and side effects. A patient can be selected and treated. Furthermore, by comparing the concentrations of VEGF-A (vascular endothelial growth factor-A, vascular endothelial growth factor-A) in the serum on the 21st day before and after treatment, the effect can be predicted more accurately at an early stage of treatment. be able to.
培養細胞株におけるレゴラフェニブ感受性を示す図。The figure which shows the regorafenib sensitivity in a cultured cell strain. 培養細胞株におけるレゴラフェニブによるVEGF‐A分泌量の変化を示す図。The figure which shows the change of VEGF-A secretion amount by the regorafenib in a cultured cell strain. 治療開始前の血清中のCCL‐5濃度と生存解析の結果を示す図。図3(A)に腫瘍縮小とCCL‐5値のROC曲線を、図3(B)に、CCL‐5≦59.96(ng/mL)、CCL‐5>59.96(ng/mL)の無増悪生存期間を、図3(C)に全生存期間を示す。The figure which shows the result of CCL-5 density | concentration in a serum before a treatment start, and a survival analysis. FIG. 3 (A) shows the ROC curve of tumor shrinkage and CCL-5 value. FIG. 3 (B) shows CCL-5 ≦ 59.96 (ng / mL), CCL-5> 59.96 (ng / mL). The progression-free survival period is shown in FIG. 3 (C). 治療開始前と開始後のVEGF‐A値の変化と無増悪生存期間を解析した図。図4(A)は、VEGF‐Aの治療開始後21日目の減少と無増悪生存期間との関係を、図4(B)はVEGF‐Aの治療開始後21日目の減少と原病増悪時での増加と無増悪生存期間との関係を示す。The figure which analyzed the change of the VEGF-A value before a treatment start, and after a start, and progression-free survival. FIG. 4 (A) shows the relationship between the decrease on day 21 after the start of VEGF-A treatment and progression-free survival, and FIG. 4 (B) shows the decrease on day 21 after the start of treatment with VEGF-A and the primary disease. The relationship between the increase in exacerbation and progression-free survival is shown. 治療開始前CCL‐5濃度と、治療開始後のVEGF‐A濃度を組み合わせて無増悪生存期間の解析を行った図。図5(A)は治療開始前CCL‐5濃度がカットオフ値以下であり、治療開始後のVEGF‐A濃度が減少する群と治療開始前CCL‐5濃度がカットオフ値より高値であり、治療開始後のVEGF‐A濃度が増加する群との比較を示す。図5(B)はさらに、両者に分類されない群を併せて示す。The figure which analyzed progression-free survival time combining CCL-5 density | concentration before treatment start and VEGF-A density | concentration after treatment start. FIG. 5 (A) shows that the CCL-5 concentration before the start of treatment is less than or equal to the cut-off value, the VEGF-A concentration after the start of treatment decreases, and the CCL-5 concentration before the start of treatment is higher than the cut-off value. The comparison with the group in which the VEGF-A density | concentration after a treatment start increases is shown. FIG. 5B further shows a group not classified into both. 治療開始前CCL‐5濃度と、治療開始後のVEGF‐A濃度を組み合わせて全生存期間の解析を行った図。図6(A)は治療開始前CCL‐5濃度がカットオフ値以下であり、治療開始後のVEGF‐A濃度が減少する群と治療開始前CCL‐5濃がカットオフ値より高値であり、治療開始後のVEGF‐A濃度が増加する群との比較を示す。図6(B)はさらに、両者に分類されない群を併せて示す。The figure which analyzed the total survival time combining the CCL-5 density | concentration before a treatment start, and the VEGF-A density | concentration after a treatment start. FIG. 6A shows that the CCL-5 concentration before the start of treatment is less than or equal to the cut-off value, the VEGF-A concentration after the start of treatment decreases, and the CCL-5 concentration before the start of treatment is higher than the cut-off value. The comparison with the group in which the VEGF-A density | concentration after a treatment start increases is shown. FIG. 6B further shows a group that is not classified into both. 治療開始前CCL‐5濃度と、治療開始後及び原病増悪期のVEGF‐A濃度を組み合わせて無増悪生存期間の解析を行った図。図7(A)は治療開始前CCL‐5濃度がカットオフ値以下であり、治療開始後のVEGF‐A濃度が減少、原病増悪期のVEGF‐A濃度が増加する群と治療開始前CCL‐5濃度がカットオフ値より高値であり、治療開始後のVEGF‐A濃度が増加、原病増悪期のVEGF‐A濃度が減少する群との比較を示す。図7(B)はさらに、両者に分類されない群を併せて示す。The figure which analyzed the progression-free survival time combining CCL-5 density | concentration before a treatment start, and the VEGF-A density | concentration after a treatment start and a disease-exacerbation stage. FIG. 7A shows a group in which the CCL-5 concentration before the start of treatment is below the cut-off value, the VEGF-A concentration after the start of treatment decreases, and the VEGF-A concentration in the exacerbation stage increases, and the CCL before the start of treatment. The comparison is made with the group in which the -5 concentration is higher than the cut-off value, the VEGF-A concentration is increased after the start of the treatment, and the VEGF-A concentration is decreased in the exacerbation stage. FIG. 7B further shows a group that is not classified into both. 治療開始前CCL‐5濃度と、治療開始後及び原病増悪期のVEGF‐A濃度を組み合わせて全生存期間の解析を行った図。図8(A)は治療開始前CCL‐5濃度がカットオフ値以下であり、治療開始後のVEGF‐A濃度が減少、原病増悪期のVEGF‐A濃度が増加する群と治療開始前CCL‐5濃度がカットオフ値より高値であり、治療開始後のVEGF‐A濃度が増加、原病増悪期のVEGF‐A濃度が減少する群との比較を示す。図8(B)はさらに、両者に分類されない群を併せて示す。The figure which analyzed the whole survival time combining the CCL-5 density | concentration before a treatment start, and the VEGF-A density | concentration after the start of treatment, and a disease-exacerbation stage. FIG. 8A shows a group in which the CCL-5 concentration before the start of treatment is below the cut-off value, the VEGF-A concentration after the start of treatment decreases, the VEGF-A concentration in the exacerbation stage increases, and the CCL before the start of treatment. The comparison is made with the group in which the -5 concentration is higher than the cut-off value, the VEGF-A concentration is increased after the start of the treatment, and the VEGF-A concentration is decreased in the exacerbation stage. FIG. 8B further shows a group not classified into both.
 本発明において、有害事象とは薬物との因果関係がはっきりしないものも含め、薬物を投与された患者に生じたあらゆる好ましくない、あるいは意図しない徴候、症状、又は病気を指し、投与する薬物との因果関係が知られている副作用に限るものではない。また、無増悪生存期間(Progression Free;Survival、PFS)、全生存期間(Overall Survival;OS)は、次のように定義する。無増悪生存期間は、投与開始日を起算日として、増悪(PD)と判断された日またはあらゆる原因(死亡原因は問わない)による死亡日のいずれか早い日までとする。増悪と判断されない生存例では、増悪がないことが確認された最終日をもって打ち切りとして取り扱う。すなわち、有害事象で中止となった時に増悪が確認されなければ打ち切りとして扱う。 In the present invention, an adverse event refers to any undesirable or unintended sign, symptom, or illness that has occurred in a patient to whom a drug has been administered, including those in which the causal relationship with the drug is not clear. It is not limited to side effects with known causal relationships. Further, progression-free survival (Progression Free; Survival, PFS) and overall survival (Overall Survival; OS) are defined as follows. The progression-free survival period is from the date of start of administration to the date on which the exacerbation (PD) is determined or the death date due to any cause (regardless of cause of death), whichever comes first. Surviving cases that are not judged to be exacerbated are treated as censored on the last day when it is confirmed that there is no exacerbation. In other words, if an exacerbation is not confirmed when the event is canceled due to an adverse event, it is treated as being terminated.
 全生存期間(OS)とは、投与開始日を起算日として、あらゆる原因による死亡日までを全生存期間とする。生存例では最終生存確認日をもって打ち切りとし、また追跡不能例では追跡不能となる前に生存が確認された最終日をもって打ち切りとして扱う。 “Overall survival period (OS)” means the total survival period from the start date of administration to the date of death due to any cause. In surviving cases, the date of confirmation of final survival is censored, and in the case of inability to follow up, the censored date is determined to be the last date of confirmation of survival before becoming untraceable.
 本発明では試料とは、血液、血漿、血清、尿、腹水、胸水をいう。本発明のマーカーがこれら試料に含まれることがすでに知られているからである(非特許文献4~8)。特に血液、血漿、血清が本発明のバイオマーカーを感度良く測定できることから試料として好ましい。治療開始前の試料とは、標準化学療法で不応または不耐となった患者に対し、レゴラフェニブによる治療を開始する前の試料を指す。また初回治療開始後早期の試料とは、実施例では21日目の試料を用いているが、1サイクル(1サイクルは3週間連日経口投与後、1週間の休薬期間を設ける。)目の試料であればいずれの時期の試料を用いてもよい。薬剤投与の効果を考慮すると、14日目~28日目の試料が好ましい。 In the present invention, the sample refers to blood, plasma, serum, urine, ascites, pleural effusion. This is because it is already known that the marker of the present invention is contained in these samples (Non-Patent Documents 4 to 8). In particular, blood, plasma, and serum are preferable as samples because the biomarker of the present invention can be measured with high sensitivity. The sample before starting treatment refers to a sample before starting treatment with regorafenib for patients who have become refractory or intolerant to standard chemotherapy. Moreover, although the sample of the 21st day is used for the sample at the early stage after the start of the first treatment in the Examples, the first cycle is one cycle (one cycle is provided with a one week rest period after oral administration for 3 weeks every day). Any sample may be used as long as it is a sample. Considering the effect of drug administration, the samples on the 14th to 28th days are preferable.
 また、ここではELISAによって、患者試料の検討を行ったが、試料中のバイオマーカーのタンパク質量を測定することができれば、どのような測定方法であってもよい。測定感度が高く、検査が比較的簡便に行えることから、イムノアッセイによって測定することが好ましい。イムノアッセイには、ELISA以外にも、例えば、ラジオイムノアッセイ(RIA)、蛍光イムノアッセイ(FIA)、蛍光偏光イムノアッセイ(FPIA)、化学発光イムノアッセイ(CLIA)等があるが、いずれの方法を用いてもよい。 In addition, although the patient sample was examined here by ELISA, any measurement method may be used as long as the protein amount of the biomarker in the sample can be measured. Measurement is preferably performed by immunoassay because measurement sensitivity is high and testing can be performed relatively easily. In addition to ELISA, for example, radioimmunoassay (RIA), fluorescence immunoassay (FIA), fluorescence polarization immunoassay (FPIA), chemiluminescence immunoassay (CLIA), and the like can be used.
 「カットオフ値」は、その値を基準として疾患の判定をした場合に、高い診断感度(有害事象率)及び高い診断特異度の両方を満足できる値である。例えば、有害事象を発症した個体で高い陽性率を示し、かつ、有害事象を発症していない個体で高い陰性率を示す値をカットオフ値として設定することが出来る。具体的なカットオフ値については、用いる試料やアッセイ系によって異なることから、各アッセイ系に応じて適宜定めることが可能である。また、カットオフ値の算出方法は、この分野において周知の方法(例えば、非特許文献9~11参照。)で定めればよい。 The “cut-off value” is a value that can satisfy both high diagnostic sensitivity (adverse event rate) and high diagnostic specificity when a disease is determined based on the value. For example, a value showing a high positive rate in an individual who has developed an adverse event and a high negative rate in an individual who has not developed an adverse event can be set as the cutoff value. Since the specific cut-off value varies depending on the sample and assay system used, it can be appropriately determined according to each assay system. The cut-off value calculation method may be determined by a method known in this field (for example, see Non-Patent Documents 9 to 11).
 本発明のキットには、本発明で見出されたバイオマーカーであるCCL‐5、VEGF‐Aを検出するための抗体、及びこれを検出するための試薬等が含むことができる。検出試薬としては、二次抗体、基質剤、標識物質(例えば、蛍光色素、酵素)が含まれる。また、これらの要素は、必要に応じてあらかじめ混合しておくこともできる。さらに、マイクロタイタープレートなどの固相、反応容器や、洗浄液や抗体を希釈するための緩衝液、陽性対照、陰性対照、プロトコールを記載した指示書などを含むことができる。 The kit of the present invention can contain an antibody for detecting CCL-5 and VEGF-A, which are biomarkers found in the present invention, a reagent for detecting this, and the like. The detection reagent includes a secondary antibody, a substrate agent, and a labeling substance (for example, fluorescent dye, enzyme). Moreover, these elements can also be mixed beforehand as needed. Further, it may include a solid phase such as a microtiter plate, a reaction container, a buffer for diluting a washing solution or an antibody, a positive control, a negative control, an instruction describing a protocol, and the like.
 以下、本発明について具体的に説明する。
(1)培養細胞を用いた解析
 最初に前臨床の実験および生物情報解析により、バイオマーカーとなる候補因子の絞り込みを行った。レゴラフェニブは、前述のようにマルチキナーゼ阻害剤であり、癌細胞自身のシグナル伝達や血管新生に関わる癌の周辺環境因子への作用が示唆されている。ただし、そのような因子の中で具体的にどの因子がレゴラフェニブ処理により直接に影響を受けるかは明確でなかった。そこで本発明者らは、血管新生に関与する因子や炎症性サイトカインから選択した候補に加え、マイクロアレイによる遺伝子発現解析やELISA法によって検出された癌細胞から分泌される因子の変動について解析を行った。
Hereinafter, the present invention will be specifically described.
(1) Analysis using cultured cells First, candidate factors as biomarkers were narrowed down by preclinical experiments and biological information analysis. Regorafenib is a multikinase inhibitor, as described above, and has been suggested to act on cancer cell environmental signals related to signal transduction and angiogenesis. However, it was not clear which of these factors was directly affected by regorafenib treatment. Therefore, the present inventors analyzed changes in factors secreted from cancer cells detected by microarray gene expression analysis or ELISA method, in addition to candidates selected from factors involved in angiogenesis and inflammatory cytokines. .
 最初に培養細胞株がレゴラフェニブに対して異なる感受性を有するか解析を行った。ヒト大腸癌細胞株として、ATCCより入手可能なHCT‐15、HCT‐116、HT‐29を用いた。 First, it was analyzed whether the cultured cell lines have different sensitivities to regorafenib. HCT-15, HCT-116 and HT-29 available from ATCC were used as human colon cancer cell lines.
 上記各大腸癌細胞株を10%FBS-RPMI培地を用い、9000個/mLの細胞密度で播種し、24時間後にレゴラフェニブを0μM、1μM、3μMの濃度になるように添加し72時間培養した。細胞増殖をMTSアッセイ(プロメガ社製)により解析した。結果を図1に示す。 The above colon cancer cell lines were seeded at a cell density of 9000 cells / mL using 10% FBS-RPMI medium, 24 hours later, regorafenib was added to a concentration of 0 μM, 1 μM, and 3 μM and cultured for 72 hours. Cell proliferation was analyzed by MTS assay (Promega). The results are shown in FIG.
 図1に示すように、いずれの細胞株もレゴラフェニブ添加によって、濃度依存的に細胞増殖が抑制される。特にHT‐29、HCT‐116細胞株は非常に強く細胞増殖が抑制され、レゴラフェニブに対する感受性が高い。一方、HCT‐15細胞株は、3μMでレゴラフェニブを添加した場合でも、他の細胞株に比べて細胞増殖が抑制される程度が少なく、レゴラフェニブに対して抵抗性を備えた細胞株であることが明らかとなった。 As shown in FIG. 1, cell growth is suppressed in a concentration-dependent manner by adding regorafenib. In particular, the HT-29 and HCT-116 cell lines are extremely strong and suppress cell growth and are highly sensitive to regorafenib. On the other hand, even when regorafenib is added at 3 μM, the HCT-15 cell line is a cell line that is less resistant to cell growth than other cell lines and has resistance to regorafenib. It became clear.
 これらレゴラフェニブに異なる感受性を有する3種の大腸癌細胞株を用いて、ELISA法によりレゴラフェニブを添加することにより癌細胞から分泌される因子の変動の解析を行った。その結果、細胞のレゴラフェニブ感受性に相関して分泌量に顕著な変化のある因子としてVEGF‐Aを見出した(図2)。 Using these three types of colorectal cancer cell lines having different sensitivities to regorafenib, changes in factors secreted from cancer cells were analyzed by adding regorafenib by ELISA. As a result, VEGF-A was found as a factor having a significant change in the secretion amount in correlation with the regorafenib sensitivity of the cells (FIG. 2).
 大腸癌細胞株HCT‐15、HCT‐116、HT‐29に濃度を変えてレゴラフェニブを添加し、48時間後に培地に分泌されているVEGF‐A濃度の測定をELISA(R&Dシステムズ社のQuantikine ELISAキット)によって解析した。各培養細胞株でレゴラフェニブを添加せずに培養した場合の培地中のVEGF‐A濃度を100%としてVEGF‐A濃度を示している。 Regorafenib was added to colorectal cancer cell lines HCT-15, HCT-116, and HT-29, and VEGF-A concentration secreted into the culture medium was measured 48 hours later by ELISA (Quantikine ELISA kit from R & D Systems) ). The VEGF-A concentration is shown with the VEGF-A concentration in the culture medium being 100% when cultured without adding regorafenib in each cultured cell line.
 レゴラフェニブ抵抗性の大腸癌細胞(HCT‐15)ではレゴラフェニブ処理後にVEGF‐Aの分泌量が上昇する。一方、レゴラフェニブ感受性の大腸癌細胞(HT‐29、HCT‐116)ではレゴラフェニブ処理後に分泌量が強く低下することが明らかになった。 In regorafenib-resistant colorectal cancer cells (HCT-15), VEGF-A secretion increases after regorafenib treatment. On the other hand, it was clarified that the amount of secretion in legolafenib-sensitive colon cancer cells (HT-29, HCT-116) was strongly reduced after treatment with regorafenib.
 VEGF‐Aの大腸癌細胞における発現については、Oncomine database(https://www.oncomine.org/resource/login.html)より情報を得ることができる。これによると、VEGF‐Aは大腸癌細胞での発現亢進が認められる。また、Kumarら(非特許文献12)によると、大腸癌患者血清では健常人血清より顕著にVEGF‐Aの濃度が高いこと、またVEGF-Aレベルは進行癌でより高いことが示されている。これらのことから、レゴラフェニブの投与対象となる進行大腸癌患者における血清VEGF‐Aは大腸癌に依存したものであることが強く示唆される。 Information on the expression of VEGF-A in colon cancer cells can be obtained from Oncomine database (https://www.oncomine.org/resource/login.html). According to this, expression of VEGF-A is increased in colon cancer cells. In addition, according to Kumar et al. (Non-patent Document 12), it is shown that the concentration of VEGF-A is significantly higher in the serum of colorectal cancer patients than in the healthy human serum, and the VEGF-A level is higher in advanced cancer. . These facts strongly suggest that serum VEGF-A in patients with advanced colorectal cancer to whom regorafenib is administered depends on colorectal cancer.
 これらの文献上の知見と得られたデータを合わせ、VEGF‐Aを中心とするサイトカインの血清レベルの変化がレゴラフェニブ感受性の良いマーカーとなりうると予測をたて、実際の患者血清を用いた前向き研究を進めた。VEGF‐Aなどに加え、IL‐8など他の血管新生に関わるサイトカインの変化や、やはり癌の悪性化に関わる炎症に関与する液性因子についても合わせて検討を行った。VEGF‐Aに加え、CCL‐2、CCL‐5、IL‐8(interleukin-8)、PlGF(placental growth factor)、Ang‐1(angiopoietin-1)、Ang‐2(angiopoietin-2)、bFGF(basic fibroblast growth factor)、SDF‐1(stromal cell-derived factor 1)、PDGFβ(platelet-derived growth factor beta)を候補因子として、患者血清を用いて解析を行うことにした。 A prospective study using actual patient sera, predicting that changes in serum levels of cytokines, especially VEGF-A, could be a good marker for regorafenib sensitivity, combining these findings with the data obtained Advanced. In addition to VEGF-A and the like, changes in other angiogenesis cytokines such as IL-8 and humoral factors also involved in inflammation related to cancer malignancy were also examined. In addition to VEGF-A, CCL-2, CCL-5, IL-8 (interleukin-8), PlGF (Placent Growth Factor), Ang-1 (angiopoietin-1), Ang-2 (angiopoietin-2), bFGF ( Analysis was performed using patient serum using basic fibroblast growth factor), SDF-1 (stromal cell-derived factor 1), and PDGFβ (platelet-derived growth factor beta) as candidate factors.
(2)患者血清を用いた解析
[対象]
 本発明では標準化学療法で不応または不耐となった進行再発結腸・直腸癌患者を対象とし、以下の選択基準によりレゴラフェニブを投与し解析を行った。
(2) Analysis using patient serum [subject]
In the present invention, patients with advanced recurrent colorectal cancer who were refractory or intolerant to standard chemotherapy were subjected to analysis by administering regorafenib according to the following selection criteria.
選択基準
1.組織学的に大腸癌と診断された症例。
2.フルオロウラシル(5‐FU)、オキサリプラチン、イリノテカン、分子標的薬剤(ベバシズマブ、セツキシマブ、パニツムマブ)を用いた標準化学療法で不応または不耐が確認された症例。
3.測定可能病変を有する症例(固形癌の治療効果判定のための新ガイドライン、New Response Evaluation Criteria in Solid Tumours;RECIST ver.1.1.に準拠)。
4.全身症状の指標であるPS(Performance Status;ECOG(Eastern Cooperative Oncology Group)基準)が0~1の症例。
5.投与開始日から12週以上の生存が期待される症例。
6.試験参加について患者本人から文書による同意が得られた症例。
Selection criteria A case histologically diagnosed as colorectal cancer.
2. Cases in which refractory or intolerance has been confirmed by standard chemotherapy using fluorouracil (5-FU), oxaliplatin, irinotecan, and molecular targeted drugs (bevacizumab, cetuximab, panitumumab).
3. Cases with measurable lesions (according to New Guideline for Evaluation of Treatment Effect for Solid Cancer, New Response Evaluation Criteria in Solid Tumors; RECIST ver.1.1).
4). Cases where PS (Performance Status; ECOG (Eastern Cooperative Oncology Group) standard), which is an index of systemic symptoms, is 0 to 1.
5). Patients who are expected to survive for 12 weeks or more from the start date of administration.
6). A case where written consent was obtained from the patient himself / herself for participation in the study.
除外基準
1.活動性の重複癌を有する症例(同時性重複癌または無病期間が5年以内の異時性重複癌。ただし局所治療により治癒と判断されるCarcinoma in situ(上皮内癌)または粘膜内癌相当の病変は活動性の重複癌に含めない)。
2.試験施行に重大な支障をきたすと考えられる合併症(間質性肺炎、肺線維症、高度の肺気腫、腸管麻痺、腸閉塞、コントロール不良な糖尿病、肝硬変、コントロール不良な高血圧症、3ヵ月以内の心筋梗塞の既往、心疾患、コントロール不良な狭心症または不整脈等)を有する症例。
3.臨床上問題となる精神・神経症状等により試験への参加が困難と判断される症例。
4.医師が登録には不適当と判断した症例。
Exclusion criteria Cases with active double cancer (simultaneous double cancer or metachronous double cancer with a disease-free period of 5 years or less. However, it is equivalent to Carcinoma in situ (localized cancer) judged to be cured by local treatment or intramucosal cancer. Lesions are not included in active double cancer).
2. Complications that may cause serious problems in the study (interstitial pneumonia, pulmonary fibrosis, severe emphysema, intestinal paralysis, bowel obstruction, uncontrolled diabetes, liver cirrhosis, uncontrolled hypertension, myocardium within 3 months Patients with a history of infarction, heart disease, poorly controlled angina or arrhythmia).
3. Cases where participation in the study is considered difficult due to clinically problematic psychiatric and neurological symptoms.
4). Cases judged by doctors to be inappropriate for registration.
 具体的には、2013年5月から2014年12月までに54名の登録患者を解析の対象とした。抗腫瘍効果は、病勢コントロール(Disease control、DC)が確認された患者は51.9%、腫瘍縮小(Tumor shrinkage、TS)が確認された患者は35.5%であった。無増悪生存期間は中央値97日、全生存期間は中央値264日であった。 Specifically, 54 registered patients were analyzed from May 2013 to December 2014. The anti-tumor effect was 51.9% for patients with confirmed disease control (Disease control, DC) and 35.5% for patients with confirmed tumor reduction (Tumor shrinkage, TS). Median progression-free survival was 97 days and overall survival was median 264 days.
 [治療法]
 レゴラフェニブを原病増悪、用量調節困難な有害事象発現まで継続する。通常、成人には1日1回レゴラフェニブ160mgを食後に3週間連日経口投与し、その後1週間休薬する。これを1サイクルとして投与を繰り返す。なお、患者の状態により適宜減量する。
[Treatment]
Continue to use regorafenib until exacerbation of the disease and onset of adverse events that are difficult to control. In general, for adults, 160 mg of regorafenib is orally administered once daily for 3 weeks after meals, and then withdrawn for 1 week. This is repeated as one cycle. The dose may be reduced according to the patient's condition.
 [検査法]
1.血清学的測定項目と測定法
 レゴラフェニブの初回治療開始前、初回治療開始後21日目、治療中止判定時に約20mLの血液を採取し、血清を分離して-80℃で保存する。候補としたサイトカインをELISAで測定した。
[Inspection method]
1. Serological measurement items and measurement method Before starting the first treatment of regorafenib, on the 21st day after the start of the first treatment, about 20 mL of blood is collected at the time of treatment discontinuation, and the serum is separated and stored at −80 ° C. Candidate cytokines were measured by ELISA.
 血清中におけるCCL‐5、VEGF‐Aなどのサイトカイン量については、R&Dシステムズ社のQuantikine ELISAキットを用いたサンドウィッチELISA法により添付のプロトコールに従い行った。 The amount of cytokines such as CCL-5 and VEGF-A in the serum was determined according to the attached protocol by the sandwich ELISA method using Quantikine ELISA kit of R & D Systems.
 まず、各サイトカインに対する抗体がコートされた96ウェル・プレートにキット付属のバッファーを加えた後、血清サンプルおよび検量線作成用のサイトカイン標品(希釈系列)を添加し、室温で2時間反応させた。その後、洗浄用バッファーにて3度洗浄の後、各サイトカインに対するHRP標識抗体を添加し、さらに室温で1-2時間反応した。その後、洗浄用バッファーにて3度洗浄の後、発色液を添加し、室温で20-25分反応後、反応停止液を加え、OD450およびOD570(バックグラウンド)を、プレートリーダーを用いて計測した。各サンプルは3連で実験を行い、バックグラウンドを引いた値について検量線に基づき、サイトカイン濃度を算出した。 First, the buffer attached to the kit was added to a 96-well plate coated with an antibody against each cytokine, and then a serum sample and a cytokine preparation for preparing a calibration curve (dilution series) were added and reacted at room temperature for 2 hours. . Thereafter, after washing three times with a washing buffer, an HRP-labeled antibody against each cytokine was added, and further reacted at room temperature for 1-2 hours. Thereafter, after washing three times with a washing buffer, a coloring solution was added, and after reacting at room temperature for 20-25 minutes, a reaction stop solution was added, and OD450 and OD570 (background) were measured using a plate reader. . Each sample was tested in triplicate, and the cytokine concentration was calculated based on the calibration curve for the value obtained by subtracting the background.
2.抗腫瘍評価、生存期間
 8週間毎にCT等の画像検査により、RECIST ver.1.1で評価する。生存期間は無増悪生存期間、全生存期間をKaplan‐Meier法を用いて算出した。
2. Anti-tumor evaluation, survival period RECIST ver. Evaluate with 1.1. Survival time was calculated using the Kaplan-Meier method.
3.有害事象の評価
 CTCAE ver.4.0(Common Terminology Criteria for Adverse Events v4.0;有害事象共通用語規準v4.0、日本語訳JCOG版)に従い重症度を分類した。
3. Assessment of adverse events CTCAE ver. The severity was classified according to 4.0 (Common Terminology Criteria for Adverse Events v4.0; Adverse Event Common Terminology Criteria v4.0, Japanese translation JCOG version).
 [解析方法]
 病勢コントロール(DC)と非病勢コントロール(Non‐DC)あるいは腫瘍縮小(TS)と非腫瘍縮小(Non‐TS)の各々2群に分けて解析を行い、抗腫瘍効果の予測因子の選定を行った。有害事象に関しては、CTCAE4.0によるグレードをもとに有害事象と相関する予測因子を選定した。さらに有害事象の発現と治療効果の相関についても解析を行った。
[analysis method]
Analyzes were conducted in two groups: disease control (DC) and non-morbidity control (Non-DC) or tumor reduction (TS) and non-tumor reduction (Non-TS), and predictors of antitumor effects were selected. It was. Regarding adverse events, predictors that correlate with adverse events were selected based on grades according to CTCAE 4.0. We also analyzed the correlation between the occurrence of adverse events and therapeutic effects.
 上述の候補因子に関して、レゴラフェニブの初回治療開始前、初回治療開始後21日目、治療中止判定時の3点でELISAを行い、各候補因子が有害事象の発症と相関を有するか解析を行った。 Regarding the above-mentioned candidate factors, ELISA was conducted at 3 points before the start of the first treatment of regorafenib, 21 days after the start of the first treatment, and at the time of the decision to stop the treatment, and whether each candidate factor was correlated with the occurrence of an adverse event was analyzed. .
 [結果]
 転移性大腸癌のレゴラフェニブ単剤療法を受けた54例について解析を行った。対象となった患者背景を表1に示す。なお、対象とした54名の患者は、2013年以降にがん研有明病院で治療を行い、インフォームドコンセントを得ている。
[result]
We analyzed 54 patients who received regorafenib monotherapy for metastatic colorectal cancer. The subject patient background is shown in Table 1. The targeted 54 patients have been treated at the Cancer Institute Ariake Hospital since 2013 and obtained informed consent.
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001
 解析したサイトカインのうち有効性、有害事象との相関が認められたのはCCL‐5とVEGF‐Aであった。試料中のCCL‐5とVEGF‐Aレベルは、有効性、及び日本人に特に頻度が高いとされる有害事象発症、すなわち、手足症候群、高血圧、肝機能異常、高ビリルビン血症、血小板減少に関する有効な予測マーカーであることが明らかとなった。以下結果について詳述する。 Among the analyzed cytokines, it was CCL-5 and VEGF-A that were correlated with efficacy and adverse events. CCL-5 and VEGF-A levels in the sample are related to efficacy and incidence of adverse events that are particularly common in Japanese, ie hand-foot syndrome, hypertension, liver dysfunction, hyperbilirubinemia, thrombocytopenia It became clear that it was an effective predictive marker. The results will be described in detail below.
 [抗腫瘍効果とサイトカインの解析]
(1)治療開始前の血清CCL‐5濃度と、腫瘍縮小、生存期間との相関
 治療開始前の血清CCL‐5値と腫瘍縮小(TS)に関するROC(receiver operating characteristic curve)解析により、CCL‐5≦59.96(ng/mL、カットオフ値)の場合に、有意に腫瘍縮小が獲得された。図3(A)は、腫瘍縮小と血清中のCCL‐5濃度についてROC曲線を作成したものである。この結果から治療開始前の血清中のCCL‐5濃度がCCL‐5≦59.96(ng/mL)であれば、レゴラフェニブ投与によって腫瘍縮小効果を期待することができる。カットオフ値は、標準曲線を用いて算出しているため、この値から大きくずれることは考えにくいが、測定法、患者の病態のグレードによって変化することが考えられる。その場合は、適宜カットオフ値を算出しなおすことが好ましい。Student’s T‐testによれば、CCL‐5 low(平均値50.5±23.8ng/mL)はCCL‐5 high(平均値65.8±23.2ng/mL)に対して、腫瘍縮小が期待できる(p=0.030)。また、生存解析でも、CCL‐5≦59.96(ng/mL、カットオフ値)で無増悪生存期間(PFS、図3(B))、全生存期間(OS、図3(C))とも統計学的に有意な改善傾向を認めた。したがって、治療開始前のCCL‐5を測定することにより、腫瘍縮小、無増悪生存期間、全生存期間を予測することが可能である。
[Anti-tumor effect and cytokine analysis]
(1) Correlation between serum CCL-5 concentration before start of treatment and tumor shrinkage, survival time By ROC (receiver operating characteristic curve) analysis on serum CCL-5 value and tumor shrinkage (TS) before start of treatment, CCL- Significant tumor reduction was obtained when 5 ≦ 59.96 (ng / mL, cut-off value). FIG. 3 (A) shows an ROC curve created for tumor shrinkage and serum CCL-5 concentration. From this result, if the CCL-5 concentration in the serum before the start of treatment is CCL-5 ≦ 59.96 (ng / mL), a tumor reducing effect can be expected by administration of regorafenib. Since the cut-off value is calculated using a standard curve, it is unlikely that the cut-off value will deviate significantly from this value, but it may vary depending on the measurement method and the patient's disease state grade. In that case, it is preferable to recalculate the cutoff value as appropriate. According to Student's T-test, CCL-5 low (mean value 50.5 ± 23.8 ng / mL) was compared to CCL-5 high (mean value 65.8 ± 23.2 ng / mL) Reduction can be expected (p = 0.030). In the survival analysis, CCL-5 ≦ 59.96 (ng / mL, cut-off value), progression-free survival (PFS, FIG. 3 (B)), and overall survival (OS, FIG. 3 (C)). A statistically significant improvement trend was observed. Therefore, it is possible to predict tumor shrinkage, progression-free survival, and overall survival by measuring CCL-5 before the start of treatment.
(2)VEGF‐A濃度の変化と生存期間との相関
 治療開始前と治療開始後のサイトカイン値の変化を解析した結果、治療前後のVEGF‐A濃度の変化と生存期間が相関することが明らかとなった。VEGF‐A濃度が、治療前より治療開始後21日目で減少した場合には、無増悪生存期間は平均146日であるのに対し、増加した場合には、無増悪生存期間は平均62日であった(図4(A))。
(2) Correlation between changes in VEGF-A concentration and survival time Analysis of changes in cytokine levels before and after treatment reveals that the change in VEGF-A concentration before and after treatment correlates with survival time It became. If the VEGF-A concentration decreases from 21 days after the start of treatment than before treatment, the progression-free survival averages 146 days, whereas if increased, the progression-free survival averages 62 days (FIG. 4A).
 また、VEGF‐A濃度が21日目に減少し、原病増悪時に増加するパターンは無増悪生存期間が改善され、無増悪生存期間の平均値は146日であった。これに対し、VEGF‐A濃度が21日目に増加し、原病増悪時に減少するパターンは無増悪生存期間が55日であった(図4(B))。治療開始後21日目のVEGF‐A濃度と、原病増悪時の濃度とは統計学的に有意な相関が見られた。また、統計学的には有意ではないが、全生存期間でも同様の傾向を示すことが認められた。すなわち、治療開始後の血清VEGF‐A値の変化が治療効果を予測する因子であることが明らかとなった。 In addition, the VEGF-A concentration decreased on the 21st day, and the pattern of increase during exacerbation of the original disease improved the progression-free survival period, and the average value of the progression-free survival period was 146 days. On the other hand, the VEGF-A concentration increased on the 21st day, and the pattern of decrease during the exacerbation of the primary disease had a progression-free survival period of 55 days (FIG. 4 (B)). There was a statistically significant correlation between the VEGF-A concentration on day 21 after the start of treatment and the concentration at the time of exacerbation of the original disease. Moreover, although it was not statistically significant, it was recognized that the same tendency was shown also in the whole survival period. That is, it became clear that the change in serum VEGF-A level after the start of treatment is a factor predicting the therapeutic effect.
(3)CCL‐5濃度、VEGF‐Aの変化と生存期間の相関
 上記の(1)及び(2)の結果から、治療開始前の血清中のCCL‐5値と治療開始後のVEGF‐A濃度の変化を組み合わせて解析した結果、さらに無増悪生存期間と全生存期間の改善が明確となった。
(3) Correlation between changes in CCL-5 concentration and VEGF-A and survival time From the results of (1) and (2) above, CCL-5 values in serum before treatment start and VEGF-A after treatment start As a result of combining the changes in concentration, further improvement in progression-free survival and overall survival was clarified.
(3‐1)治療開始前CCL‐5濃度と治療開始後21日目のVEGF‐A濃度との組み合せ解析
 治療開始前のCCL‐5濃度がカットオフ値以下であり、治療開始後21日目のVEGF‐A濃度が減少した患者群(Good群)と、その対極である治療開始前のCCL‐5濃度がカットオフ値より高く、治療開始後21日目のVEGF‐A濃度が増加している患者群(Poor群)とを比較した。その結果、Good群では、有意に無増悪生存期間が改善していることが明らかとなった(図5(A))。
(3-1) Combination analysis of CCL-5 concentration before treatment start and VEGF-A concentration on the 21st day after the start of treatment The CCL-5 concentration before the start of the treatment is below the cut-off value, and the 21st day after the start of the treatment The group of patients with reduced VEGF-A concentration (Good group) and its opposite CCL-5 concentration before the start of treatment was higher than the cutoff value, and the VEGF-A concentration on the 21st day after the start of treatment increased. The patient group (Poor group) was compared. As a result, it was revealed that the progression-free survival period was significantly improved in the Good group (FIG. 5A).
 また、上記に分類されない患者群をMedium群として解析した。Kaplan‐Meier曲線から、Medium群の無増悪生存期間はGood群、Poor群の中間に位置する結果であった(図5(B))。Good群、Medium群、Poor群の無増悪生存期間の平均値は、それぞれ188日、69日、70日であった。 Moreover, the patient group which is not classified into the above was analyzed as a Medium group. From the Kaplan-Meier curve, the progression-free survival period of the Medium group was a result intermediate between the Good group and the Poor group (FIG. 5B). Mean values of progression-free survival in the Good group, Medium group, and Poor group were 188 days, 69 days, and 70 days, respectively.
 さらに、全生存期間について解析を行った。上記と同様に治療開始前のCCL‐5濃度がカットオフ値以下であり、治療開始後21日目のVEGF‐A濃度が減少した患者群(Good群)と、その対極である治療開始前のCCL‐5濃度がカットオフ値より高く、治療開始後21日目のVEGF‐A濃度が増加している患者群(Poor群)とを比較した(図6(A))。また、どちらにも属さない患者群をMedium群として全生存期間を比較した(図6(B))。全生存期間についても無増悪生存期間と同様の傾向を示した。全生存期間の平均値はGood群が414日であるのに対し、Medium群では187日、Poor群では239日であった。 Furthermore, an analysis was performed on the overall survival time. As described above, the CCL-5 concentration before the start of treatment was below the cut-off value, and the VEGF-A concentration decreased on the 21st day after the start of treatment (Good group), and the opposite electrode before the start of treatment A comparison was made with a patient group (Poor group) in which the CVEGF-5 concentration was higher than the cut-off value and the VEGF-A concentration increased on the 21st day after the start of treatment (FIG. 6 (A)). Moreover, the patient group which does not belong to either was made into the Medium group, and the whole survival time was compared (FIG.6 (B)). The overall survival was similar to progression-free survival. The mean overall survival was 414 days in the Good group, compared to 187 days in the Medium group and 239 days in the Poor group.
(3‐2)治療開始前CCL‐5濃度と治療開始後21日目のVEGF‐A濃度、原病増悪期のVEGF‐A濃度の組み合せ解析
 治療開始前のCCL‐5濃度がカットオフ値以下であり、治療開始後21日目のVEGF‐A濃度が減少し、さらに原病増悪期のVEGF‐A濃度が増加している患者群(Good群)と、その対極である治療開始前のCCL‐5濃度がカットオフ値より高く、治療開始後21日目のVEGF‐A濃度が増加し、さらに原病増悪期のVEGF‐A濃度が減少している患者群(Poor群)とを比較した(図7(A))。その結果、Good群では有意に無増悪生存期間が改善されていることが明らかであった。また、これら以外をMedium群として解析した。その結果、Kaplan‐Meier曲線から、Medium群の無増悪生存期間はその中間の結果であった(図7(B))。Good群、Medium群、Poor群の無増悪生存期間の平均値は、それぞれ232日、70日、53日であった。
(3-2) Combined analysis of CCL-5 concentration before the start of treatment and VEGF-A concentration on the 21st day after the start of treatment and VEGF-A concentration in the stage of exacerbation of the disease CCL-5 concentration before the start of treatment is below the cut-off value A group of patients (Good group) in which the VEGF-A concentration on the 21st day after the start of treatment is decreased and the VEGF-A concentration in the stage of exacerbation of disease is increased, and the CCL before the start of the treatment, which is the opposite electrode Compared with the patient group (Poor group) in which the VEGF-A concentration was higher than the cutoff value, the VEGF-A concentration increased on the 21st day after the start of treatment, and the VEGF-A concentration in the exacerbation period was decreased (FIG. 7 (A)). As a result, it was clear that the progression-free survival period was significantly improved in the Good group. Other than these were analyzed as a Medium group. As a result, from the Kaplan-Meier curve, the progression-free survival period of the Medium group was an intermediate result (FIG. 7B). Mean values of progression-free survival in the Good group, Medium group, and Poor group were 232 days, 70 days, and 53 days, respectively.
 さらに、全生存期間について解析を行った。上記と同様に治療開始前のCCL‐5濃度がカットオフ値以下であり、治療開始後21日目のVEGF‐A濃度が減少し、さらに原病増悪期のVEGF‐A濃度が増加している患者群(Good群)と、その対極である治療開始前のCCL‐5濃度がカットオフ値より高く、治療開始後21日目のVEGF‐A濃度が増加し、さらに原病増悪期のVEGF‐A濃度が減少している患者群(Poor群)とを比較した(図8(A))。また、どちらにも属さない群をMedium群として全生存期間を比較した(図8(B))。全生存期間についても無増悪生存期間と同様の傾向を示した。全生存期間の平均値はGood群が414日であるのに対し、Medium群では239日、Poor群では192日であった。 Furthermore, an analysis was performed on the overall survival time. Similar to the above, the CCL-5 concentration before the start of treatment is below the cut-off value, the VEGF-A concentration on the 21st day after the start of treatment decreased, and the VEGF-A concentration during the exacerbation stage increased The patient group (Good group) and its opposite CCL-5 concentration before the start of treatment were higher than the cut-off value, the VEGF-A concentration increased on the 21st day after the start of the treatment, and VEGF- A comparison was made with a patient group (Poor group) in which the A concentration decreased (FIG. 8 (A)). Moreover, the group which does not belong to neither was made into the Medium group, and the whole survival time was compared (FIG.8 (B)). The overall survival was similar to progression-free survival. The mean overall survival was 414 days in the Good group, compared to 239 days in the Medium group and 192 days in the Poor group.
 以上の結果から、血清CCL‐5とVEGF‐Aは単独または併用することでレゴラフェニブ単剤療法の治療前あるいは治療開始早期の効果予測マーカーとして用いることができる。 From the above results, serum CCL-5 and VEGF-A can be used alone or in combination as a marker for predicting the effect of regorafenib monotherapy before treatment or at the beginning of treatment.
(4)有害事象とサイトカインの解析
 有害事象の発症と治療開始前のサイトカイン血清値との間に相関があるか解析を行った。有害事象の評価項目は、レゴラフェニブで特に頻度が高い手足症候群、高血圧、肝機能異常、高ビリルビン血症、血小板減少を選択した。有害事象と相関の高いサイトカイン濃度を表2に示す。
(4) Analysis of adverse events and cytokines An analysis was conducted to determine whether there was a correlation between the onset of adverse events and serum levels of cytokines before the start of treatment. The endpoints for adverse events were regorafenib, the most common hand-foot syndrome, hypertension, liver dysfunction, hyperbilirubinemia, and thrombocytopenia. Table 2 shows cytokine concentrations that are highly correlated with adverse events.
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002
 表中、AEは有害事象(Adverse Event)、T‐Bilは総ビリルビン量を示す。ASTはアスパラギン酸アミノトランスフェラーゼ、ALTはアラニンアミノトランスフェラーゼの略称であり、ともに肝機能の指標となる検査値である。また、HTは高血圧(hypertension)、HFSは手足症候群(hand‐foot syndrome)を表す。また、Ang‐2、bFGF、CCL‐2は上記のとおりである。 In the table, AE indicates an adverse event (Adverse Event), and T-Bil indicates the total amount of bilirubin. AST is an abbreviation for aspartate aminotransferase and ALT is an abbreviation for alanine aminotransferase, both of which are test values serving as indicators of liver function. HT represents hypertension, and HFS represents hand-foot syndrome. Ang-2, bFGF, and CCL-2 are as described above.
 治療開始前のサイトカイン血清値と有害事象との間に統計学的に有意な差を認めたのは、Ang‐2、bFGF、CCL‐2の値であった。Ang‐2が高値の場合に高ビリルビン血症(grade3≦)が多かった。また、bFGF高値でAST(grade3≦)、ALT(grade3≦)が高く、bFGFと肝機能の相関が見られた。また、bFGF低値で高血圧(grade3≦)が多く、またCCL‐2低値で手足症候群(grade2≦)が多かった。これらの有害事象の発症は治療中の休薬や減量に大きく関わるものである。治療開始前にこれらサイトカインの濃度を測定することにより、特定の有害事象の発症を予測し得ることにより、より注意深く患者の状態をモニターしながらレゴラフェニブを投薬することができる。すなわち、治療開始前のAng‐2、bFGF、CCL‐2濃度は、治療遂行に関与する有害事象発症の予測マーカーとなり得る。 It was the values of Ang-2, bFGF, and CCL-2 that showed statistically significant differences between cytokine serum levels before the start of treatment and adverse events. When Ang-2 was high, hyperbilirubinemia (grade 3 ≦) was high. Moreover, AST (grade3 ≦) and ALT (grade3 ≦) were high at high bFGF, and a correlation between bFGF and liver function was observed. In addition, bFGF was low and hypertension (grade 3 ≦) was high, and CCL-2 was low and limb syndrome (grade 2 ≦) was high. The occurrence of these adverse events is largely related to withdrawal and weight loss during treatment. By measuring the concentrations of these cytokines before the start of treatment, one can predict the onset of a specific adverse event, so that regorafenib can be dosed while monitoring the patient's condition more carefully. That is, Ang-2, bFGF, and CCL-2 concentrations before the start of treatment can be predictive markers for the onset of adverse events involved in treatment performance.
 また、レゴラフェニブの抗腫瘍効果を判定する際に有用とされた治療開始前CCL‐5≦59.96(ng/mL、カットオフ値)と有害事象についてカイ二乗検定により解析を行った。結果を表3に示す。なお、表中PLTは血小板減少、ORはオッズ比を示し、他の略号は表2と同様である。 Further, CCL-5 ≦ 59.96 (ng / mL, cut-off value) before starting treatment and adverse events, which were useful in determining the antitumor effect of regorafenib, were analyzed by chi-square test. The results are shown in Table 3. In the table, PLT represents thrombocytopenia, OR represents an odds ratio, and other abbreviations are the same as those in Table 2.
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003
 CCL‐5濃度がカットオフ値(CO)以下の場合、手足症候群(grade1≦)と血小板減少(grade1≦)の発症が有意に多かった。手足症候群(HFS)グレード1≦は、CCL-5がカットオフ値以下の場合9.1倍発症しやすく、血小板減少(PLT)グレード1≦は、約4.2倍発症しやすいという結果が得られた。しかし、重篤例の発症には相関しなかった。そのほか、高ビリルビン血症(grade2≦)もやや高頻度であった。 When the CCL-5 concentration was below the cut-off value (CO), the onset of hand-foot syndrome (grade1 ≦) and thrombocytopenia (grade1 ≦) were significantly higher. Hand-foot syndrome (HFS) grade 1 ≦ is 9.1 times more likely to develop when CCL-5 is below the cut-off value, and thrombocytopenia (PLT) grade 1 ≦ is about 4.2 times more likely to occur. It was. However, it did not correlate with the onset of serious cases. In addition, hyperbilirubinemia (grade 2 ≦) was slightly more frequent.
 さらに、治療前後でのCCL‐5濃度の変化、治療開始前のCCL‐5濃度(CO値以下)と治療開始後のVEGF‐A濃度の変化の組み合わせ、あるいは治療開始後21日目のVEGF濃度の変化と有害事象との相関をカイ二乗検定により解析した。結果を表4に示す。 Further, change in CCL-5 concentration before and after treatment, combination of CCL-5 concentration before treatment start (CO value or less) and change in VEGF-A concentration after treatment start, or VEGF concentration on the 21st day after treatment start The correlation between changes in AEs and adverse events was analyzed by chi-square test. The results are shown in Table 4.
Figure JPOXMLDOC01-appb-T000004
Figure JPOXMLDOC01-appb-T000004
 治療前後でのCCL‐5濃度の変化と有害事象との相関では、治療後にCCL‐5濃度が低下した場合に、特にgrade2≦のAST値が相関し肝機能異常発症が多かった。治療開始前のCCL‐5濃度(CO値以下)と治療開始後のVEGF‐A濃度の変化の組み合わせは、高ビリルビン血症(grade2≦)と血小板減少(grade1≦、grade2≦)の発症に関与していることが明らかとなった。また、VEGF単独でも同様の結果を得ている。すなわち治療開始後21日目のVEGF‐A濃度が低下する場合には、高ビリルビン血症(grade1≦)、高ビリルビン血症(grade2≦)、血小板減少(grade2≦)が有意に高頻度であった。 Regarding the correlation between changes in CCL-5 concentration before and after treatment and adverse events, AST values of grade 2 ≦ were particularly correlated when CCL-5 concentration decreased after treatment, and liver function abnormalities were frequently developed. Combination of CCL-5 concentration (below CO value) before treatment start and change in VEGF-A concentration after treatment start is associated with the development of hyperbilirubinemia (grade2 ≦) and thrombocytopenia (grade1 ≦, grade2 ≦) It became clear that Similar results were obtained with VEGF alone. That is, when the VEGF-A concentration on the 21st day after the start of treatment decreased, hyperbilirubinemia (grade1 ≦), hyperbilirubinemia (grade2 ≦), and thrombocytopenia (grade2 ≦) were significantly more frequent. It was.
 以上の結果から、CCL‐5とVEGF‐Aはサルベージ療法におけるレゴラフェニブ単剤療法において日本人で発症率の高い有害事象発症の治療前あるいは治療開始早期の効果予測マーカーとして用いることができる。 From the above results, CCL-5 and VEGF-A can be used as predictive markers for the effect of pretreatment or early treatment onset of adverse events with high incidence in Japanese in regorafenib monotherapy in salvage therapy.
 また、これら有害事象とCCL‐5、VEGF‐Aとの相関は、無増悪生存期間、全生存期間との相関に矛盾するものではない。治療前のCCL‐5濃度、治療早期のVEGF‐A濃度をレゴラフェニブの有効性の指標として使用し、さらに上記CCL‐5、VEGF‐A濃度と相関を有する有害事象に関して注意深く観察しながら、患者の治療を行えばよい。また、併せて治療開始前のAng‐2、bFGF、CCL‐2濃度を測定することによって、有害事象を予測することができる。 Also, the correlation between these adverse events and CCL-5 and VEGF-A is not inconsistent with the correlation between progression-free survival and overall survival. Using the CCL-5 concentration before treatment and the VEGF-A concentration at the early stage of treatment as an indicator of the effectiveness of regorafenib, and carefully observing the adverse events correlated with the CCL-5 and VEGF-A concentrations, Treatment can be performed. Moreover, an adverse event can be predicted by measuring the Ang-2, bFGF, and CCL-2 concentrations before starting treatment.
 本発明ではレゴラフェニブについて詳細に解析を行ったが、化合物の構造ないし活性が類似するマルチキナーゼ阻害剤であるソラフェニブ、スニチニブ等の他のマルチキナーゼ阻害剤への適用も期待できる。 In the present invention, regorafenib was analyzed in detail, but application to other multikinase inhibitors such as sorafenib and sunitinib which are multikinase inhibitors having similar structures and activities of the compounds can also be expected.
 本発明で示したように、抗がん剤投与、化学療法によく反応する患者を選択して治療を行うことにより、治療の有効な患者にのみ抗がん剤治療を行い、不要な治療や重篤な副作用が生じるリスクを避けることが可能となる。 As shown in the present invention, anticancer drug treatment is performed only on patients who are effective in treatment by selecting patients who respond well to anticancer drug administration and chemotherapy. The risk of serious side effects can be avoided.

Claims (13)

  1.  レゴラフェニブ(Regorafenib)投与による効果を予測するための検査方法であって、
     患者より採取された試料中のCCL‐5濃度を測定することを特徴とする検査方法。
    A test method for predicting the effect of administration of regorafenib,
    A test method comprising measuring CCL-5 concentration in a sample collected from a patient.
  2.  請求項1記載の検査方法であって、
     前記試料が血液、血漿、血清、尿、腹水、胸水であることを特徴とする検査方法。
    The inspection method according to claim 1,
    The test method, wherein the sample is blood, plasma, serum, urine, ascites, or pleural effusion.
  3.  請求項1又は2記載の検査方法であって、
     前記試料が治療開始前のものであることを特徴とする検査方法。
    The inspection method according to claim 1 or 2,
    An inspection method, wherein the sample is one before the start of treatment.
  4.  請求項3記載の検査方法であって、
     CCL‐5濃度が所定値より低い場合には、
     無増悪生存期間(PFS)及び全生存期間(OS)の改善が期待できると判定する検査方法。
    The inspection method according to claim 3,
    If the CCL-5 concentration is lower than the specified value,
    A test method for determining that improvement in progression-free survival (PFS) and overall survival (OS) can be expected.
  5.  請求項1、2、又は4のいずれか1項記載の検査方法であって、
     患者試料中の治療開始前と初回治療開始後早期のVEGF‐A濃度を測定し、
     治療開始後のVEGF‐A濃度が治療開始前と比較して減少傾向にある場合には、
     PFS及びOSの改善が期待できると判定する検査方法。
    The inspection method according to any one of claims 1, 2, or 4,
    Measure the VEGF-A concentration in the patient sample before the start of treatment and early after the start of the first treatment,
    When the VEGF-A concentration after the start of treatment is decreasing compared to before the start of treatment,
    Inspection method for determining that improvement of PFS and OS can be expected.
  6.  請求項5記載の検査方法であって、
     原病増悪時の患者試料中のVEGF‐A濃度を測定し、
     初回治療開始後早期のVEGF‐A濃度と比較して増加している場合には、
     PFS及びOSの改善が期待できると判定する検査方法。
    The inspection method according to claim 5,
    Measure VEGF-A concentration in patient sample at the time of exacerbation
    If there is an increase compared to the early VEGF-A concentration after initial treatment,
    Inspection method for determining that improvement of PFS and OS can be expected.
  7.  請求項1~6のいずれか1項記載の検査方法であって、
     治療開始前の患者試料中のAng‐2、bFGF、CCL‐2のうち少なくとも1つを測定し、
     有害事象が生じるリスクを判定する検査方法。
    The inspection method according to any one of claims 1 to 6,
    Measuring at least one of Ang-2, bFGF, CCL-2 in a patient sample prior to treatment;
    A test method to determine the risk of adverse events.
  8.  レゴラフェニブ投与による効果を予測するための検査キットであって、
     患者試料中のCCL‐5濃度を測定するための試薬を含むことを特徴とする検査キット。
    A test kit for predicting the effects of regorafenib administration,
    A test kit comprising a reagent for measuring CCL-5 concentration in a patient sample.
  9.  請求項8記載の検査キットであって、
     さらに、VEGF‐A濃度を測定するための試薬を含むことを特徴とする検査キット。
    The test kit according to claim 8, wherein
    Furthermore, a test kit comprising a reagent for measuring a VEGF-A concentration.
  10.  請求項8又は9記載の検査キットであって、
     Ang‐2、bFGF、CCL‐2のうち少なくとも1つの濃度を測定するための試薬を含むことを特徴とするキット。
    The test kit according to claim 8 or 9, wherein
    A kit comprising a reagent for measuring the concentration of at least one of Ang-2, bFGF and CCL-2.
  11.  請求項8~10いずれか1項記載のキットであって、
     前記試薬がイムノアッセイ用の試薬であることを特徴とするキット。
    A kit according to any one of claims 8 to 10,
    A kit, wherein the reagent is an immunoassay reagent.
  12.  レゴラフェニブ投与による効果を予測するためのバイオマーカーであって、
     CCL‐5及び/又はVEGF‐Aであることを特徴とするバイオマーカー。
    A biomarker for predicting the effects of regorafenib administration,
    A biomarker characterized by being CCL-5 and / or VEGF-A.
  13.  レゴラフェニブ投与による有害事象を予測するためのバイオマーカーであって、
     CCL‐5、VEGF‐A、Ang‐2、bFGF、CCL‐2の少なくとも1つからなることを特徴とするバイオマーカー。
    A biomarker for predicting adverse events due to regorafenib administration,
    A biomarker comprising at least one of CCL-5, VEGF-A, Ang-2, bFGF, and CCL-2.
PCT/JP2016/087859 2015-12-24 2016-12-20 Test method for predicting effectiveness and safety of multi-kinase inhibitor, test kit, and biomarker WO2017110764A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2017558133A JP6853789B2 (en) 2015-12-24 2016-12-20 Test methods, test kits, and biomarkers that predict the efficacy and safety of multikinase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2015251386 2015-12-24
JP2015-251386 2015-12-24

Publications (1)

Publication Number Publication Date
WO2017110764A1 true WO2017110764A1 (en) 2017-06-29

Family

ID=59090395

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2016/087859 WO2017110764A1 (en) 2015-12-24 2016-12-20 Test method for predicting effectiveness and safety of multi-kinase inhibitor, test kit, and biomarker

Country Status (2)

Country Link
JP (1) JP6853789B2 (en)
WO (1) WO2017110764A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011146725A1 (en) * 2010-05-19 2011-11-24 Bayer Healthcare Llc Biomarkers for a multikinase inhibitor
JP2014521070A (en) * 2011-06-29 2014-08-25 アムジェン インコーポレイテッド Predictive biomarkers of survival in the treatment of renal cell carcinoma
WO2014148557A1 (en) * 2013-03-19 2014-09-25 凸版印刷株式会社 Method for predicting sensitivity to egfr inhibitor
WO2015031604A1 (en) * 2013-08-28 2015-03-05 Crown Bioscience, Inc. Gene expression signatures predictive of subject response to a multi-kinase inhibitor and methods of using the same
JP2015527874A (en) * 2012-05-31 2015-09-24 バイエル ファーマ アクチエンゲゼルシャフト Biomarkers for determining an effective response to treatment of hepatocellular carcinoma (HCC) patients

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011146725A1 (en) * 2010-05-19 2011-11-24 Bayer Healthcare Llc Biomarkers for a multikinase inhibitor
JP2014521070A (en) * 2011-06-29 2014-08-25 アムジェン インコーポレイテッド Predictive biomarkers of survival in the treatment of renal cell carcinoma
JP2015527874A (en) * 2012-05-31 2015-09-24 バイエル ファーマ アクチエンゲゼルシャフト Biomarkers for determining an effective response to treatment of hepatocellular carcinoma (HCC) patients
WO2014148557A1 (en) * 2013-03-19 2014-09-25 凸版印刷株式会社 Method for predicting sensitivity to egfr inhibitor
WO2015031604A1 (en) * 2013-08-28 2015-03-05 Crown Bioscience, Inc. Gene expression signatures predictive of subject response to a multi-kinase inhibitor and methods of using the same

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ANNA KUBO: "Regorafenib ni yoru Kan Shogai Akka Yosoku Kanosei no Kent o", ANNUAL MEETING OF JAPANESE SOCIETY OF MEDICAL ONCOLOGY, vol. 12 th, 19 July 2014 (2014-07-19), pages 3 - 10-3 *
AZUSA KOMORI: "Regorafenib no Soki Koka Yosoku wa Kano ka? Shizuoka Gan.Aichi Gan Kyodo Kansatsu Kenkyu [2]", THE ANNUAL MEETING OF JAPAN SOCIETY OF CLINICAL ONCOLOGY, vol. 53 rd, 29 October 2015 (2015-10-29), pages 124 - 1 *
MASAHIDE FUKUDO: "Studies on biomarker related to toxicity of molecular-targeted anticancer drugs toward safe and individualized therapy", ADVANCES IN PHARMACEUTICAL SCIENCES, vol. 32, 1 March 2016 (2016-03-01), pages 109 - 114 *
NAO KAKIZAWA: "Shinko Saihatsu Daichogan ni Taisuru Regorafenib Tekio Handan no Shihyo", ANNUAL CONGRESS OF JAPAN SURGICAL SOCIETY, vol. 116 th, 14 April 2016 (2016-04-14), pages 064 - 5 *
SHIN'YAKU, NEWS [ NIPPON HYOJUN SHOHIN BUNRUI BANGO] 874291 REGORAFENIB-JO (STIVARGA-JO® 40MG, vol. 62, no. 4, 31 August 2013 (2013-08-31), pages 327 - 329 *
SHOTA FUKUOKA: "Regorafenib no Biomarker", MOLECULAR TARGETED THERAPY FOR CANCE R, vol. 12, no. 2, 20 June 2014 (2014-06-20), pages 210 - 215 *
SHOZO OSERA: "Daichogan no Saishin Chiryo I. Soron Perspective of personalized (precise) cancer therapy based on predictive biomarkers", JAPANESE JOURNAL OF CLINICAL MEDICINE, vol. 72, no. 1, 1 January 2014 (2014-01-01), pages 29 - 34 *
SUENAGA MITSUKUNI: "Serum VEGF-A and CCL5 levels as candidate biomarkers for efficacy and toxicity of regorafenib in patients with metastatic colorectal cancer", ONCOTARGET, vol. 7, no. 23, 7 June 2016 (2016-06-07), pages 34811 - 34823, XP055396494 *

Also Published As

Publication number Publication date
JPWO2017110764A1 (en) 2018-11-01
JP6853789B2 (en) 2021-03-31

Similar Documents

Publication Publication Date Title
US9315869B2 (en) Marker for predicting gastric cancer prognosis and method for predicting gastric cancer prognosis using the same
JP2015503920A (en) Biomarkers for breast cancer prediction and diagnosis
AU2017301970B2 (en) Combination test for colorectal cancer
WO2012151574A1 (en) Methods of developing a prognosis for pancreatic cancer and predicting responsiveness to cancer therapeutics
JP2013538337A (en) Plasma biomarkers for bevacizumab combination therapy for the treatment of pancreatic cancer
US20240103014A1 (en) Inspection Method Enabling Specific Diagnosis of Pathological State of Diabetic Nephropathy at Early Stage
WO2021116057A1 (en) Biomarker panel for diagnosing colorectal cancer
WO2011122634A1 (en) Prognostic method for pulmonary adenocarcinoma, pulmonary adenocarcinoma detection kit, and pharmaceutical composition for treating pulmonary adenocarcinoma
Duan et al. Correlation among genetic variations of c-MET in Chinese patients with non-small cell lung cancer
US11499973B2 (en) Methods of predicting responsiveness of a cancer to a VEGF targeting agent and methods of prognosing and treating cancer
WO2017110764A1 (en) Test method for predicting effectiveness and safety of multi-kinase inhibitor, test kit, and biomarker
US11415584B2 (en) Biomarkers, test method, and test kit for predicting therapeutic effect of anti-VEGFR-2 antibody drug
Sakamoto et al. Development of quantitative detection assays for CYR61 as a new marker for benign prostatic hyperplasia
EP2895863B1 (en) New biomarkers for the diagnosis and/or prognosis of clear cell renal cell carcinoma
CN109642257B (en) Method for predicting effect of drug therapy on cancer
Faulkner et al. Tumour markers
KR20190051744A (en) An anticancer agent for treating cancers resistant to EGFR inhibitor
EP2702409B1 (en) CXCR1 as a predictor of response to treatment with epidermal growth factor receptor targeted therapeutic
WO2012113819A1 (en) Molecular biomarkers for predicting response to antitumor treatment in lung cancer
US20220082565A1 (en) The fgf19-cholestyramine (f-cme) test as a two-stage method for routine cancer screening
WO2011058164A1 (en) Molecular biomarkers for predicting response to tyrosine kinase inhibitors in lung cancer
WO2015144184A1 (en) Use of timp-1 as a biomarker in the egf-receptor inhibitor treatment of metastatic colorectal cancer
US8557532B2 (en) Diagnosis and treatment of drug-resistant Ewing'S sarcoma

Legal Events

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

Ref document number: 16878653

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2017558133

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16878653

Country of ref document: EP

Kind code of ref document: A1