TW202339754A - Genomic loss of heterozygosity as a predictive biomarker for treatment with talazoparib and methods of treatment thereof - Google Patents

Genomic loss of heterozygosity as a predictive biomarker for treatment with talazoparib and methods of treatment thereof Download PDF

Info

Publication number
TW202339754A
TW202339754A TW112100665A TW112100665A TW202339754A TW 202339754 A TW202339754 A TW 202339754A TW 112100665 A TW112100665 A TW 112100665A TW 112100665 A TW112100665 A TW 112100665A TW 202339754 A TW202339754 A TW 202339754A
Authority
TW
Taiwan
Prior art keywords
cancer
gloh
talazopanib
treatment
pharmaceutically acceptable
Prior art date
Application number
TW112100665A
Other languages
Chinese (zh)
Inventor
艾倫 道格拉斯 拉爾德
Original Assignee
美商輝瑞大藥廠
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 美商輝瑞大藥廠 filed Critical 美商輝瑞大藥廠
Publication of TW202339754A publication Critical patent/TW202339754A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Abstract

The present invention relates to a method of selecting a subject having a cancer with a deficiency in homologous recombination repair, for treatment with talazoparib, or a pharmaceutically acceptable salt thereof, and methods of treatment thereof, including: a) determining a gLOH score from a biopsy of the cancer; b) selecting the subject for treatment with talazoparib, or a pharmaceutically acceptable salt thereof, based on the gLOH score; and c) administering a therapeutically effective amount of talazoparib, or a pharmaceutically acceptable salt thereof, to the selected subject.

Description

以他拉唑帕尼治療之作為預測性生物標記之基因雜合性缺失及其治療方法Genetic loss of heterozygosity as a predictive biomarker treated with talazopanib and its treatment method

本發明係關於選擇病患以用他拉唑帕尼治療之方法及其治療方法。The present invention relates to methods of selecting patients for treatment with talazopanib and methods of treatment thereof.

聚(ADP-核糖)聚合酶(PARP)參與細胞中之脫氧核醣核酸(DNA)修復的天然存在之過程。已顯示PARP抑制藉由誘導合成致死而成為針對與雙股DNA修復基因中之突變相關聯之腫瘤的一種有效治療策略(Sonnenblick, A.等人, Nat. Rev. Clin. Oncol, 2015, 12(1), 27-4)。PARP抑制在具有直接或間接涉及同源重組修復(HRR)之DNA損傷反應(DDR)基因中之同型接合缺失或有害改變或其兩者之細胞中係合成致死的(Lord, CJ等人, Science, 2017; 355: 1152-1158)。Poly(ADP-ribose) polymerase (PARP) is involved in the naturally occurring process of deoxyribonucleic acid (DNA) repair in cells. PARP inhibition has been shown to be an effective therapeutic strategy against tumors associated with mutations in double-stranded DNA repair genes by inducing synthetic lethality (Sonnenblick, A. et al., Nat. Rev. Clin. Oncol, 2015, 12( 1), 27-4). PARP inhibition is synthetically lethal in cells with homozygous deletions or deleterious alterations, or both, in DNA damage response (DDR) genes that are directly or indirectly involved in homologous recombination repair (HRR) (Lord, CJ et al., Science , 2017; 355: 1152-1158).

DDR係已演進以修復受損DNA之路徑的網路。此等尤其包括錯配修復、鹼基切除修復及同源重組修復(HRR)。鑒於HRR在修復雙股DNA斷裂中之高保真度,其在維持基因體完整性方面尤其重要。PARP之抑制導致單股DNA斷裂之累積及導致因PARP捕獲之DNA應力,此最終造成雙股DNA斷裂。因此,PARP抑制劑選擇性地致死缺失HRR之癌細胞-此為合成致死之一實例,一種基因或基因產物之功能中之缺陷單獨時具有極小作用但與第二基因或基因產物之功能中的缺陷合併時具有毒性的機制。DDR is a network of pathways that has evolved to repair damaged DNA. These include, inter alia, mismatch repair, base excision repair and homologous recombination repair (HRR). Given the high fidelity of HRR in repairing double-stranded DNA breaks, it is particularly important in maintaining genome integrity. Inhibition of PARP leads to the accumulation of single-stranded DNA breaks and leads to DNA stress trapped by PARP, which ultimately results in double-stranded DNA breaks. Thus, PARP inhibitors selectively kill cancer cells lacking HRR - an example of synthetic lethality, a defect in the function of one gene or gene product that alone has a minimal effect but is not related to a defect in the function of a second gene or gene product. Mechanisms by which defects become toxic when merged.

他拉唑帕尼係經口有效的強效小分子PARP抑制劑,其對攜帶損害DNA修復之基因突變的人類癌細胞株具有細胞毒性,該作用稱為合成致死,且藉由捕獲DNA上之PARP蛋白質來藉此防止DNA修復、複製及轉錄。Talazopanib is an orally available and potent small molecule PARP inhibitor. It is cytotoxic to human cancer cell lines carrying genetic mutations that impair DNA repair. This effect is called synthetic lethality and occurs by capturing DNA on the DNA. PARP proteins prevent DNA repair, replication and transcription.

化合物他拉唑帕尼係「(8 S,9 R)-5-氟-8-(4-氟苯基)-9-(1-甲基-1 H-1,2,4-三唑-5-基)-8,9-二氫-2 H-吡啶并[4,3,2- de]呔𠯤-3(7 H)-酮」,亦稱為「(8 S,9 R)-5-氟-8-(4-氟苯基)-9-(1-甲基-1 H-1,2,4-三唑-5-基)-2,7,8,9-四氫-3 H-吡啶并[4,3,2- de]呔𠯤-3-酮」(亦稱作「PF-06944076」、「MDV3800」及「BMN673」),係一種PARP抑制劑,具有以下結構: 他拉唑帕尼。 The compound talazopanib is "(8 S ,9 R )-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1 H -1,2,4-triazole- 5-yl)-8,9-dihydro- 2H -pyrido[4,3,2- de ]pyrido-3( 7H )-one", also known as "( 8S , 9R )- 5-Fluoro-8-(4-fluorophenyl)-9-(1-methyl-1 H -1,2,4-triazol-5-yl)-2,7,8,9-tetrahydro- 3H -pyrido[4,3,2- de ]pyrido-3-one" (also known as "PF-06944076", "MDV3800" and "BMN673") is a PARP inhibitor with the following structure: Talazopanib.

他拉唑帕尼及其醫藥學上可接受之鹽,包括甲苯磺酸鹽,揭示於國際公開案第WO 2010/017055號及第WO 2012/054698號中。製備他拉唑帕尼及其醫藥學上可接受之鹽,包括甲苯磺酸鹽之其他方法描述於國際公開案第WO 2011/097602號、第WO 2015/069851號及第WO 2016/019125號中。使用他拉唑帕尼及其醫藥學上可接受之鹽,包括甲苯磺酸鹽,治療癌症之其他方法揭示於國際公開案第WO 2011/097334號及第WO 2017/075091號中。Talazopanib and its pharmaceutically acceptable salts, including tosylate, are disclosed in International Publication Nos. WO 2010/017055 and WO 2012/054698. Other methods of preparing talazopanib and its pharmaceutically acceptable salts, including the tosylate salt, are described in International Publication Nos. WO 2011/097602, WO 2015/069851 and WO 2016/019125 . Other methods of treating cancer using talazopanib and its pharmaceutically acceptable salts, including tosylate, are disclosed in International Publication Nos. WO 2011/097334 and WO 2017/075091.

TALZENNA ®(他拉唑帕尼) (0.25 mg及1 mg膠囊)已在包括美國之若干國家及在歐盟審批通過,且在其他國家審批或在預料之批准之情況下復核以用於患有有害或疑似有害gBRCAm HER2陰性局部晚期或轉移性乳癌之成年病患的治療。0.5 mg及0.75 mg之其他膠囊濃度已在美國審批通過。他拉唑帕尼在攜帶直接或間接與HRR相關聯之基因改變的轉移性去勢抗性前列腺癌中已顯示活性(de Bono等人, Lancet Oncol. 2021年9月;22(9):1250-1264)。正在研發他拉唑帕尼作為單一藥劑及與其他藥劑合併用於各種人類癌症。 TALZENNA ® (talazopanib) (0.25 mg and 1 mg capsules) is approved in several countries, including the United States, and in the European Union, and is approved or under review in anticipation of approval in other countries for use in patients with harmful or treatment of adult patients with locally advanced or metastatic breast cancer with suspected deleterious gBRCAm HER2-negative breast cancer. Other capsule strengths of 0.5 mg and 0.75 mg have been approved in the United States. Talazopanib has shown activity in metastatic castration-resistant prostate cancer harboring genetic alterations directly or indirectly associated with HRR (de Bono et al., Lancet Oncol. 2021 Sep;22(9):1250- 1264). Talazopanib is being developed for various human cancers as a single agent and in combination with other agents.

大部分癌症藥物在一些病患中有效,但在其他病患中無效。此可歸因於腫瘤之間的基因變異,且甚至可在同一病患內之腫瘤當中觀測到。對於靶向療法,可變病患回應尤其明顯。因此,靶向療法之全部潛能在沒有適合測試用於判定哪些病患將受益於哪些藥物之情況下可能無法實現。根據國立衛生研究院(NIH),術語「生物標記(biomarker)」定義為「經客觀量測且評估為正常生物或致病過程或對治療性干預之藥理學反應之指標的一個特徵」。Most cancer drugs are effective in some patients but not others. This can be attributed to genetic variation between tumors and can even be observed among tumors within the same patient. Variable patient responses are particularly evident for targeted therapies. Therefore, the full potential of targeted therapies may not be realized without suitable tests to determine which patients will benefit from which drugs. According to the National Institutes of Health (NIH), the term "biomarker" is defined as "a characteristic that is objectively measured and assessed as an indicator of normal biological or pathogenic processes or pharmacological response to a therapeutic intervention."

存在三種不同類型之癌症生物標記:(1)預後生物標記、(2)預測性生物標記及(3)藥效學生物標記。預後生物標記用於根據攻擊性,亦即生長速率及/或轉移速率及對治療的頑抗性(refractiveness to treatment)來對癌症,例如實體腫瘤進行分類。此有時稱為區分「良好結果」腫瘤與「不良結果」腫瘤。預測性生物標記用於評定特定病患將從以特定藥物治療獲益之機率。例如,患有ERBB2 (HER2或NEU)基因經擴增之乳癌的病患可能受益於用曲妥珠單抗(HERCEPTIN ®)治療,而無ERBB2基因擴增之病患不大可能受益於用曲妥珠單抗治療。藥效學生物標記係當病患服用某種藥物時該藥物對病患之作用的一個指示。因此,在新藥物之臨床研發的早期階段期間,藥效學生物標記通常用於引導劑量水準及給藥頻率。關於癌症生物標記之論述,參見例如Sawyers, 2008, Nature452:548-552。 There are three different types of cancer biomarkers: (1) prognostic biomarkers, (2) predictive biomarkers, and (3) pharmacodynamic biomarkers. Prognostic biomarkers are used to classify cancers, such as solid tumors, according to their aggressiveness, ie, growth rate and/or metastasis rate, and refractiveness to treatment. This is sometimes called distinguishing "good outcome" tumors from "poor outcome" tumors. Predictive biomarkers are used to assess the likelihood that a specific patient will benefit from treatment with a specific drug. For example, patients with breast cancer with amplification of the ERBB2 (HER2 or NEU) gene may benefit from treatment with trastuzumab ( HERCEPTIN® ), whereas patients without ERBB2 gene amplification are unlikely to benefit from treatment with trastuzumab (HERCEPTIN®). Tocilizumab treatment. A pharmacodynamic biomarker is an indication of how a drug affects a patient when the patient takes it. Therefore, during the early stages of clinical development of new drugs, pharmacodynamic biomarkers are often used to guide dosage levels and dosing frequency. For a discussion of cancer biomarkers, see, for example, Sawyers, 2008, Nature 452:548-552.

他拉唑帕尼在攜帶DNA修復基因突變之轉移性去勢抗性前列腺癌及在gBRCA突變HER2陰性局部晚期或轉移性乳癌中具有活性;然而,他拉唑帕尼對癌症病患之效用可能超過此等適應症。此外,對於他拉唑帕尼在其中已顯示活性之適應症,並非所有病患均受益於他拉唑帕尼治療。因此,需要選擇攜帶具有DNA修復基因突變之腫瘤的哪些病患可能對他拉唑帕尼反應且需要鑑別除了患有DNA修復基因突變之癌症之病患之外可能對他拉唑帕尼治療反應的癌症病患。因此,需要基於預測性生物標記之診斷方法,該方法可用於鑑別可能(或不大可能)對以他拉唑帕尼治療反應之癌症病患。Talazopanib is active in metastatic castration-resistant prostate cancer harboring DNA repair gene mutations and in gBRCA-mutated HER2-negative locally advanced or metastatic breast cancer; however, talazopanib may be more effective in cancer patients than Such indications. In addition, not all patients benefit from treatment with talazopanib in indications in which talazopanib has shown activity. Therefore, there is a need to select which patients with tumors harboring DNA repair gene mutations are likely to respond to talazopanib and to identify patients who may respond to talazopanib treatment in addition to patients with cancers with DNA repair gene mutations. of cancer patients. Therefore, there is a need for diagnostic methods based on predictive biomarkers that can be used to identify cancer patients who are likely (or unlikely) to respond to treatment with talazopanib.

本發明部分地提供選擇病患及鑑別癌症以用他拉唑帕尼或其醫藥學上可接受之鹽治療之方法及其治療方法。提供此發明內容係以簡化形式引入下文實施方式中進一步描述之一系列的概念。此發明內容並不意欲鑑別所主張標的物的關鍵特徵或基本特徵,亦不意欲單獨地用作判定所主張標的物之範圍的輔助。The present invention provides, in part, methods of selecting patients and identifying cancers for treatment with talazopanib or a pharmaceutically acceptable salt thereof, and methods of treating the same. This Summary is provided to introduce a selection of concepts in a simplified form that are further described below in the Detailed Description. This Summary is not intended to identify key features or essential features of the claimed subject matter, nor is it intended to be used solely as an aid in determining the scope of the claimed subject matter.

根據本發明之實施例1,提供一種選擇患有具有同源重組修復缺陷之癌症之個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療之方法,其包含:a)根據癌症之活檢判定基因雜合性缺失(gLOH)分數;及b)基於gLOH分數選擇個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療。According to Embodiment 1 of the present invention, there is provided a method of selecting individuals suffering from cancer with homologous recombination repair defects for treatment with talazopanib or a pharmaceutically acceptable salt thereof, which includes: a) according to the cancer The biopsy determines the genetic loss of heterozygosity (gLOH) score; and b) selecting individuals for treatment with talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score.

根據本發明之實施例2,提供一種治療個體內具有同源重組修復缺陷之癌症之方法,其包含:a)根據癌症之活檢判定基因雜合性缺失(gLOH)分數;b)基於gLOH分數選擇個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療;及c)基於gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。According to Embodiment 2 of the present invention, a method for treating cancer with homologous recombination repair defects in an individual is provided, which includes: a) determining a gene loss of heterozygosity (gLOH) score based on a biopsy of the cancer; b) selecting based on the gLOH score The subject is treated with talazopanib or a pharmaceutically acceptable salt thereof; and c) administering a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score.

根據本發明之實施例3,提供一種鑑別對以他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,其包含:a)根據癌症之活檢判定基因雜合性缺失(gLOH)分數;及b)基於gLOH分數選擇癌症以用他拉唑帕尼或其醫藥學上可接受之鹽治療。According to Embodiment 3 of the present invention, a method for identifying cancers with homologous recombination repair defects that are sensitive to treatment with talazopanib or a pharmaceutically acceptable salt thereof is provided, which method includes: a) based on a biopsy of the cancer Determining a genetic loss of heterozygosity (gLOH) score; and b) selecting cancers for treatment with talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score.

根據本發明之實施例4,提供一種對以他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,其包含:a)根據癌症之活檢判定基因雜合性缺失(gLOH)分數;b)基於gLOH分數選擇癌症以用他拉唑帕尼或其醫藥學上可接受之鹽治療;及c)基於gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。According to Embodiment 4 of the present invention, a method for cancer with homologous recombination repair deficiency that is sensitive to treatment with talazopanib or a pharmaceutically acceptable salt thereof is provided, which method includes: a) based on biopsy determination of the cancer Gene loss of heterozygosity (gLOH) score; b) selecting cancers for treatment with talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score; and c) administering a therapeutically effective amount of talazopanib based on the gLOH score Pany or its pharmaceutically acceptable salt.

下文描述本發明之實施例,其中為方便起見,實施例1、2、3及4 (E1、E2、E3及E4)與上文所提供之實施例一致。Embodiments of the present invention are described below, wherein for convenience, Embodiments 1, 2, 3 and 4 (E1, E2, E3 and E4) are consistent with the embodiments provided above.

應理解,前文一般描述與以下詳細說明僅舉例說明及解釋而非限制所主張之本發明。It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not limiting of the invention as claimed.

參考本發明之以下實施例及較佳實施例之詳細描述,可更容易地理解本發明。應理解,本文中所使用之術語係出於僅描述具體實施例之目的且並非意欲為限制性的。應進一步理解,除非在本文中加以具體限制,否則本文中所使用之術語具有其在相關技術中所知的傳統含義。The present invention can be more easily understood with reference to the following detailed description of the embodiments and preferred embodiments of the invention. It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. It is further understood that unless specifically limited herein, terms used herein have their conventional meanings as known in the relevant art.

E1 一種選擇患有具有同源重組修復缺陷之癌症之個體之方法,如上所定義。E1 A method of selecting individuals with cancers that are defective in homologous recombination repair, as defined above.

E2 一種治療個體內之具有同源重組修復缺陷之癌症的方法,如上所定義。E2 A method of treating cancer in an individual that is defective in homologous recombination repair, as defined above.

E3 一種鑑別對用他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,如上文所定義。E3 A method of identifying cancers with homologous recombination repair deficiency that are susceptible to treatment with talazopanib or a pharmaceutically acceptable salt thereof, as defined above.

E4 一種治療對用他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,如上文所定義。E4 A method of treating cancer having homologous recombination repair deficiency that is susceptible to treatment with talazopanib or a pharmaceutically acceptable salt thereof, as defined above.

E5 如實施例1或2之方法,其中癌症對以他拉唑帕尼治療敏感。E5 The method of embodiment 1 or 2, wherein the cancer is sensitive to treatment with talazopanib.

E6 如實施例1至4中任一項之方法,其中癌症係前列腺癌。E6 The method of any one of embodiments 1 to 4, wherein the cancer is prostate cancer.

E7 如實施例6之方法,其中前列腺癌係轉移性前列腺癌。E7 The method of embodiment 6, wherein the prostate cancer is metastatic prostate cancer.

E8 如實施例6之方法,其中前列腺癌係去勢抗性前列腺癌。E8 The method of embodiment 6, wherein the prostate cancer is castration-resistant prostate cancer.

E9 如實施例8之方法,其中去勢抗性前列腺癌係轉移性去勢抗性前列腺癌。E9 The method of embodiment 8, wherein the castration-resistant prostate cancer is metastatic castration-resistant prostate cancer.

E10    如實施例1或3之方法,其中癌症之同源重組修復缺陷藉由下一代定序判定。E10: The method of embodiment 1 or 3, wherein the homologous recombination repair defect of the cancer is determined by next-generation sequencing.

E11    如實施例1或3之方法,其中步驟a)係藉由下一代定序進行。E11: The method of embodiment 1 or 3, wherein step a) is performed by next-generation sequencing.

E12    如實施例1至11中任一項之方法,其中gLOH分數係至少約8.0%。E12 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least about 8.0%.

E13    如實施例1至11中任一項之方法,其中gLOH分數係至少約8.3%。E13 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least about 8.3%.

E14    如實施例1至11中任一項之方法,其中gLOH分數係至少約8.8%。E14 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least about 8.8%.

E15    如實施例1至11中任一項之方法,其中gLOH分數係至少約9%;至少約9.2%;至少約10%;至少約11%;至少約12%;至少約13%;至少約14%;至少約15%;至少約16%;至少約17%;至少約18%;至少約19%;至少約20%;至少約21%;至少約22%;至少約23%;至少約24%;或至少約25%。E15 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least about 9%; at least about 9.2%; at least about 10%; at least about 11%; at least about 12%; at least about 13%; at least about 14%; at least about 15%; at least about 16%; at least about 17%; at least about 18%; at least about 19%; at least about 20%; at least about 21%; at least about 22%; at least about 23%; at least about 24%; or at least about 25%.

E16    如實施例1至11中任一項之方法,其中gLOH分數係至少8.0%。E16 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least 8.0%.

E17    如實施例1至11中任一項之方法,其中gLOH分數係至少8.3%。E17 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least 8.3%.

E18    如實施例1至11中任一項之方法,其中gLOH分數係至少8.8%。E18 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least 8.8%.

E19    如實施例1至11中任一項之方法,其中gLOH分數係至少9%;至少9.2%;至少10%;至少11%;至少12%;至少13%;至少14%;至少15%;至少16%;至少17%;至少18%;至少19%;至少20%;至少21%;至少22%;至少23%;至少24%;或至少25%。E19 The method of any one of embodiments 1 to 11, wherein the gLOH fraction is at least 9%; at least 9.2%; at least 10%; at least 11%; at least 12%; at least 13%; at least 14%; at least 15%; At least 16%; at least 17%; at least 18%; at least 19%; at least 20%; at least 21%; at least 22%; at least 23%; at least 24%; or at least 25%.

E20    如實施例1或2之方法,其中個體係人類。E20: The method of embodiment 1 or 2, wherein the individual system is human.

本文中所描述之本發明之各實施例可與本文中所描述之本發明之一或多個其他實施例合併,該等實施例與其所合併之實施例不一致。另外,下文描述本發明之各實施例設想本發明之化合物之醫藥學上可接受之鹽在其範圍內。Each embodiment of the invention described herein may be combined with one or more other embodiments of the invention described herein that are inconsistent with the embodiment with which it is merged. In addition, the various examples of the invention described below contemplate pharmaceutically acceptable salts of the compounds of the invention within their scope.

除非另外規定,否則如本文中所使用,單數形式「一(a/an)」及「該(the)」包括複數個參考物。Unless otherwise specified, as used herein, the singular forms "a/an" and "the" include plural references.

如本文中所使用,術語「約(about)」當用於修飾某一經數值定義之參數(例如他拉唑帕尼之量)時意謂該參數可相對於該參數之所陳述數值低或高最多10%變化。例如,約1 mg之劑量可在0.9 mg與1.1 mg之間變化。As used herein, the term "about" when used to modify a numerically defined parameter (e.g., the amount of talazopanib) means that the parameter may be lower or higher relative to the stated numerical value of the parameter. Maximum 10% variation. For example, a dose of approximately 1 mg may vary between 0.9 mg and 1.1 mg.

除非另外指明,否則如本文中所使用,「異常細胞生長(Abnormal cell growth)」係指不依賴於正常調節機制(例如喪失接觸抑制)的細胞生長。異常細胞生長可為良性(非癌性)或惡性(癌性)的。Unless otherwise specified, as used herein, "abnormal cell growth" refers to cell growth that does not rely on normal regulatory mechanisms (eg, loss of contact inhibition). Abnormal cell growth can be benign (noncancerous) or malignant (cancerous).

出於本發明之目的,「DDR突變(DDR mutation(s))」、「DDR改變(DDR alteration(s))」、「HRR突變(HRR mutation(s))」及「HRR改變(HRR alteration(s))」係指直接地或間接地在同源重組修復(HRR)中涉及之基因中的改變/突變。儘管並不像片語「DNA損傷反應(DNA damage response)」一樣科學性強,但通常應理解,「DDR」亦可稱作「DNA損傷修復」或「DNA修復」。For the purposes of the present invention, "DDR mutation(s)", "DDR alteration(s)", "HRR mutation(s)" and "HRR alteration(s)" s))” refers to changes/mutation in genes involved directly or indirectly in homologous recombination repair (HRR). Although it is not as scientific as the phrase "DNA damage response", it is generally understood that "DDR" can also be called "DNA damage repair" or "DNA repair".

術語「癌症(cancer)」、「癌性(cancerous)」及「惡性(malignant)」係指或描述哺乳動物中典型地以不受調控之細胞生長為特徵的生理學病狀。如本文中所使用,「癌症」係指由異常細胞生長引起之任何惡性及/或侵襲性生長或腫瘤。如本文中所使用,「癌症」係指根據形成腫瘤之細胞類型命名的實體腫瘤、或血液、骨髓或淋巴系統之癌症。實體腫瘤之實例包括但不限於肉瘤及癌瘤。血液癌症之實例包括但不限於白血病、淋巴瘤及骨髓瘤。術語「癌症」包括但不限於源自身體特定部位之原發性癌症、自開始之位置擴散至身體其他部分之轉移性癌症、初始原發性癌症在緩解後復發及作為個人之新原發性癌症之第二原發性癌症,該個人之先前癌症病史與後一癌症之類型不同。癌症之實例包括但不限於癌瘤、淋巴瘤、白血病、母細胞瘤及肉瘤。此類癌症之更特定實例包括鱗狀細胞癌瘤、骨髓瘤、肺癌、小細胞肺癌、小細胞前列腺癌、非小細胞肺癌、神經膠瘤、霍奇金氏淋巴瘤(hodgkin's lymphoma)、非霍奇金氏淋巴瘤(non-hogkin's lymphoma)、濾泡性淋巴瘤(FL)、彌漫性大B細胞淋巴瘤(DLCBCL)、急性骨髓白血病(AML)、多發性骨髓瘤、腸胃(道)癌、腎癌、卵巢癌、子宮癌、子宮內膜癌、肝癌、腎癌、腎細胞癌瘤、前列腺癌、去勢敏感性前列腺癌(CSPC)、去勢抗性前列腺癌(CRPC)、甲狀腺癌、黑色素瘤、軟骨肉瘤、神經母細胞瘤、胰臟癌、膠質母細胞癌、多形性細胞瘤、子宮頸癌、直腸癌、腦癌、胃癌、膀胱癌、肝癌、肝細胞癌瘤、乳癌、大腸癌、頭頸癌及唾液腺癌。The terms "cancer", "cancerous" and "malignant" refer to or describe a physiological condition in mammals that is typically characterized by unregulated cell growth. As used herein, "cancer" refers to any malignant and/or invasive growth or tumor caused by abnormal cell growth. As used herein, "cancer" refers to a solid tumor, named after the type of cells that form the tumor, or a cancer of the blood, bone marrow, or lymphatic system. Examples of solid tumors include, but are not limited to, sarcomas and carcinomas. Examples of blood cancers include, but are not limited to, leukemia, lymphoma, and myeloma. The term "cancer" includes, but is not limited to, primary cancers that originate in a specific part of the body, metastatic cancers that spread from the initial location to other parts of the body, initial primary cancers that recur after remission, and new primary cancers that are new to an individual. A second primary cancer in which the individual has a prior history of cancer that is of a different type than the subsequent cancer. Examples of cancer include, but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma. More specific examples of such cancers include squamous cell carcinoma, myeloma, lung cancer, small cell lung cancer, small cell prostate cancer, non-small cell lung cancer, glioma, hodgkin's lymphoma, non-Hodgkin's lymphoma, Non-hogkin's lymphoma (non-hogkin's lymphoma), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLCBCL), acute myeloid leukemia (AML), multiple myeloma, gastrointestinal (tract) cancer, Kidney cancer, ovarian cancer, uterine cancer, endometrial cancer, liver cancer, kidney cancer, renal cell carcinoma, prostate cancer, castration-sensitive prostate cancer (CSPC), castration-resistant prostate cancer (CRPC), thyroid cancer, melanoma , chondrosarcoma, neuroblastoma, pancreatic cancer, glioblastoma, polymorphic cell tumor, cervical cancer, rectal cancer, brain cancer, stomach cancer, bladder cancer, liver cancer, hepatocellular carcinoma, breast cancer, colorectal cancer , head and neck cancer and salivary gland cancer.

術語「病患(patient)」或「個體(subject)」係指任何需要療法或正參與臨床試驗、流行病研究或用作對照之單一個體,包括人類及哺乳動物獸醫學病患,諸如牛、馬、犬及貓。在某些較佳實施例中,病患或個體係人類。The term "patient" or "subject" refers to any single individual in need of treatment or who is participating in a clinical trial, epidemiological study or control, including human and mammalian veterinary patients, such as cattle, Horses, dogs and cats. In certain preferred embodiments, the patient or subject is a human.

如本文中所使用,術語「治療(treat/treating)」癌症意謂向患有癌症或經診斷患有癌症之個體投與根據本發明之療法以實現至少一種積極治療作用,諸如減少癌細胞數目、減小腫瘤尺寸、降低癌細胞浸潤至周邊器官中之速率、或降低腫瘤轉移或腫瘤生長之速率、逆轉、緩解、抑制此類術語所應用之病症或病狀或此類病症或病狀之一或多個症狀的進展或預防其復發。除非另外指示,否則如本文中所使用之術語「治療」係指如緊接上文所定義之「治療」般的治療行為。術語「治療」亦包括對個體之輔助治療及新輔助治療。出於本發明之目的,有益或所期望的臨床結果包括但不限於以下之一或多者:減少贅生性或癌性細胞的增殖(或破壞贅生性或癌性細胞);抑制轉移或贅生性細胞;縮小或減小腫瘤之尺寸;癌症之緩解;減輕癌症引起之症狀;提高患有癌症者的生活品質;減少治療癌症所需之其他藥物的劑量;延緩癌症之進展;治癒癌症;克服癌症之一或多種抗性機制;及延長患有癌症之病患的存活期。可以多種方法量測癌症中之積極治療作用(參見例如W. A. Weber, J. Nucl. Med.50:1S-10S (200))。在一些實施例中,藉由本發明之方法實現之治療係以下之任一者:部分反應(PR)、完全反應(CR)、疾病穩定(SD)、疾病進展(PD)、總體反應(OR)、客觀反應率(ORR)、無進展存活期(PFS)、放射PFS、無疾病存活期(DFS)及總體存活期(OS)。PFS (亦稱為「腫瘤進展時間」)指示在癌症不生長之治療期間及治療之後的時長,且包括病患已經歷CR或PR之時間量以及病患已經歷疾病穩定(SD)之時間量。DFS係指在病患保持無疾病之治療期間及治療之後的時長。OS係指與新的或未經治療之個體或病患相比預期壽命之延長。在一些實施例中,對本發明方法之反應係PR、CR、SD、PD、PFS、DFS、ORR、OR或OS之任一者。使用實體腫瘤反應評估標準版本1.1(RECIST 1.1)反應標準評定對本發明之方法的反應,包括軟組織反應之持續時間。在一些實施例中,藉由本發明之方法實現之治療藉由PSA進展時間、至開始細胞毒素化學療法的時間及具有PSA反應大於或等於50%之病患比例來量測。用於有效治療癌症病患之本發明之方法的治療方案可根據諸如以下之因素而變化:疾病病況、病患之年齡及體重,及療法在個體中引發抗癌反應之能力。雖然本發明之任一態樣的實施例可能在各個體中不能有效達成積極治療作用,但其應在如藉由此項技術中已知之任何統計檢驗所測定的統計學上顯著數目之個體中達成如此效果,該等檢驗諸如但不限於科克倫-曼特爾-亨塞爾(Cochran-Mantel-Haenszel)對數等級檢定、司圖頓t檢驗(Student's t-test)、卡方檢驗(chi2-test)、根據曼及惠特尼(Mann and Whitney)之U檢驗、克拉斯卡-瓦立斯檢驗(Kruskal-Wallis test)(H檢驗)、喬卡契爾-特波斯特拉檢驗(Jonckheere-Terpstrat-test)及威爾康秩和檢驗(Wilcon on-test)。術語「治療」亦涵蓋藉由試劑、診斷劑、結合化合物或藉由另一細胞進行之例如細胞之活體外及離體治療。 As used herein, the term "treating" cancer means administering to an individual suffering from cancer or diagnosed with cancer a therapy according to the present invention to achieve at least one positive therapeutic effect, such as reducing the number of cancer cells , reduce the size of a tumor, reduce the rate of infiltration of cancer cells into surrounding organs, or reduce the rate of tumor metastasis or tumor growth, reverse, alleviate, inhibit the disease or condition to which such terms apply, or the effect of such disease or condition Progression or prevention of recurrence of one or more symptoms. Unless otherwise indicated, the term "treatment" as used herein refers to the act of treating as "treatment" is defined immediately above. The term "treatment" also includes adjuvant and neoadjuvant treatment of an individual. For the purposes of the present invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of neoplastic or cancerous cells (or destroying neoplastic or cancerous cells); inhibiting metastasis or neoplastic cells cells; shrink or reduce the size of tumors; remission of cancer; reduce symptoms caused by cancer; improve the quality of life of people with cancer; reduce the dosage of other drugs needed to treat cancer; slow the progression of cancer; cure cancer; overcome cancer one or more resistance mechanisms; and prolonging the survival of patients with cancer. Active therapeutic effects in cancer can be measured in a variety of ways (see, eg, WA Weber, J. Nucl. Med. 50:1S-10S (200)). In some embodiments, treatment achieved by the methods of the invention is any of the following: partial response (PR), complete response (CR), stable disease (SD), progressive disease (PD), overall response (OR) , objective response rate (ORR), progression-free survival (PFS), radiation PFS, disease-free survival (DFS) and overall survival (OS). PFS (also known as "time to tumor progression") refers to the time during and after treatment when the cancer is not growing, and includes the amount of time the patient has experienced CR or PR and the amount of time the patient has experienced stable disease (SD) quantity. DFS refers to the length of time during and after treatment that a patient remains disease-free. OS refers to the increase in life expectancy compared to a new or untreated individual or patient. In some embodiments, the response to the methods of the invention is any of PR, CR, SD, PD, PFS, DFS, ORR, OR, or OS. Response to the methods of the invention, including duration of soft tissue response, was assessed using Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1) response criteria. In some embodiments, treatment achieved by the methods of the invention is measured by time to PSA progression, time to initiation of cytotoxic chemotherapy, and proportion of patients with a PSA response greater than or equal to 50%. The treatment regimen of the methods of the present invention for effective treatment of cancer patients can vary depending on factors such as the condition of the disease, the age and weight of the patient, and the ability of the therapy to elicit an anti-cancer response in the individual. Although embodiments of any aspect of the invention may not be effective in achieving a positive therapeutic effect in each individual, they should be in a statistically significant number of individuals as determined by any statistical test known in the art. To achieve such an effect, tests such as but not limited to Cochran-Mantel-Haenszel log-rank test, Student's t-test, chi-square test (chi2 -test), U test based on Mann and Whitney, Kruskal-Wallis test (H test), Jonckheere test -Terpstrat-test) and Wilcon on-test. The term "treatment" also encompasses in vitro and ex vivo treatment of, for example, a cell by means of an agent, diagnostic agent, binding compound or by another cell.

如本文中所使用,藥物、化合物或醫藥調配物之「劑量(dosage)」、「量(amount)」、「有效劑量(effective dosage)」或「有效量(effective amount)」係足以對疾病、其併發症及在疾病發展期間呈現之中間病理性表型中之任何一或多種症狀(生物化學、組織學及/或行為)具有有益或所需作用的量。對於治療用途,「治療有效量(therapeutically effective amount)」係指將在一定程度上緩解所治療的病症之症狀中之一或多者的化合物投與量。關於癌症之治療,治療有效量係指具有以下作用的量:(1)減小腫瘤之尺寸,(2)抑制(即在一定程度上減緩,較佳地停止)腫瘤轉移,(3)在某種程度上抑制(即在某種程度上減緩,較佳地停止)腫瘤生長或腫瘤侵襲性,(4)在某種程度上減輕(或較佳地消除)與癌症相關聯之一或多種病徵或症狀,(5)減少治療疾病所需之其他藥物的劑量,(6)增強另一種藥物之作用,及/或(7)延緩病患之疾病進展。有效劑量可以一或多種投與形式投與。出於本發明之目的,藥物、化合物或醫藥調配物之有效劑量為足以直接或間接實現預防性或治療性治療之量。如在臨床情形下所理解,藥物、化合物或醫藥調配物之有效劑量可或可不連同另一藥物、化合物或醫藥調配物一起實現。As used herein, a "dosage", "amount", "effective dosage" or "effective amount" of a drug, compound or pharmaceutical formulation is sufficient to treat a disease, An amount that has a beneficial or desirable effect on any one or more symptoms (biochemical, histological and/or behavioral) of its complications and intermediate pathological phenotypes present during the development of the disease. For therapeutic uses, a "therapeutically effective amount" means an amount of a compound administered that will alleviate to some extent one or more of the symptoms of the condition being treated. Regarding the treatment of cancer, a therapeutically effective amount refers to an amount that has the following effects: (1) reduce the size of the tumor, (2) inhibit (i.e., slow down to a certain extent, preferably stop) tumor metastasis, (3) (i.e., slow down, preferably stop) tumor growth or tumor invasiveness to a certain extent, (4) alleviate (or preferably eliminate) one or more symptoms associated with cancer to a certain extent or symptoms, (5) reduce the dose of other drugs needed to treat the disease, (6) enhance the effect of another drug, and/or (7) delay the progression of the patient's disease. An effective dose may be administered in one or more administration forms. For the purposes of the present invention, an effective dose of a drug, compound or pharmaceutical formulation is an amount sufficient to effect, directly or indirectly, prophylactic or therapeutic treatment. As understood in the clinical context, an effective dose of a drug, compound or pharmaceutical formulation may or may not be achieved together with another drug, compound or pharmaceutical formulation.

在一個實施例中,一定量之他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以一日一次約0.1 mg至約2 mg、較佳一日一次約0.25 mg至約1.5 mg、且更佳一日一次約0.5 mg至約1.0 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.1 mg、約0.25 mg、約0.35 mg、約0.5 mg、約0.75 mg或約1.0 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.1 mg、約0.25 mg、約0.35 mg或約0.5 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.25 mg、約0.35 mg或約0.5 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.5 mg、約0.75 mg或約1.0 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.1mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.25 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.35 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.5 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約0.75 mg之每日劑量投與。在一個實施例中,他拉唑帕尼或其醫藥學上可接受之鹽且較佳其甲苯磺酸鹽以每日一次約1.0 mg之每日劑量投與。本文所提供之劑量係指游離鹼形式之他拉唑帕尼之劑量,或按經投與之他拉唑帕尼鹽形式之游離鹼當量計算。例如,他拉唑帕尼之劑量或量,諸如0.5、0.75 mg或1.0 mg係指游離鹼當量。可調整此劑量方案以提供最佳治療性反應。例如,劑量可如由治療情況之緊急狀態指示按比例減少或增加。In one embodiment, a certain amount of talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate is administered once a day at about 0.1 mg to about 2 mg, preferably at a dosage of about 0.25 mg once a day. mg to about 1.5 mg, and preferably administered at a daily dose of about 0.5 mg to about 1.0 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate is administered once daily at about 0.1 mg, about 0.25 mg, about 0.35 mg, about 0.5 mg, about 0.75 mg mg or a daily dose of approximately 1.0 mg administered. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate is administered once daily at about 0.1 mg, about 0.25 mg, about 0.35 mg or about 0.5 mg once daily. Dosage administration. In one embodiment, talazopanib, or a pharmaceutically acceptable salt thereof and preferably its tosylate salt, is administered at a daily dose of about 0.25 mg, about 0.35 mg, or about 0.5 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate salt is administered at a daily dose of about 0.5 mg, about 0.75 mg, or about 1.0 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate salt is administered at a daily dose of about 0.1 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate salt is administered at a daily dose of about 0.25 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate salt is administered at a daily dose of about 0.35 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate salt is administered at a daily dose of about 0.5 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate salt is administered at a daily dose of about 0.75 mg once daily. In one embodiment, talazopanib or a pharmaceutically acceptable salt thereof and preferably its tosylate salt is administered at a daily dose of about 1.0 mg once daily. The dosages provided herein refer to the dosage of the free base form of talazopanib or calculated as the free base equivalent of the salt form of talazopanib administered. For example, doses or amounts of talazopanib such as 0.5, 0.75 mg or 1.0 mg refer to free base equivalents. This dosage regimen can be adjusted to provide optimal therapeutic response. For example, the dosage may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.

除非另外指明,否則如本文中所使用之術語「醫藥學上可接受之鹽(pharmaceutically acceptable salt)」係指對其所投與之生物體不產生顯著刺激且不消除化合物之生物活性及特性的化合物之調配物。在某些情況下,醫藥學上可接受之鹽係藉由使本文中所描述之化合物與酸,諸如鹽酸、氫溴酸、硫酸、硝酸、磷酸、甲烷磺酸、乙烷磺酸、對甲苯磺酸、水楊酸及其類似物反應而獲得。在一些情況下,醫藥學上可接受之鹽藉由使本文中所描述之具有酸性基團之化合物與鹼反應以形成鹽或藉由預先確定之其他方法來獲得,該鹽諸如銨鹽;鹼金屬鹽,諸如鈉鹽或鉀鹽;鹼土金屬鹽,諸如鈣鹽或鎂鹽;有機鹼之鹽,諸如二環己胺;N-甲基-D-還原葡糖胺;參(羥甲基)甲胺及與諸如精胺酸、離胺酸及其類似物之胺基酸的鹽。Unless otherwise specified, the term "pharmaceutically acceptable salt" as used herein refers to salts that do not cause significant irritation to the organism to which they are administered and do not eliminate the biological activity and properties of the compound. Formulations of compounds. In some cases, pharmaceutically acceptable salts are prepared by combining a compound described herein with an acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methane sulfonic acid, ethane sulfonic acid, p-toluene Obtained from the reaction of sulfonic acid, salicylic acid and their analogues. In some cases, pharmaceutically acceptable salts are obtained by reacting a compound having an acidic group described herein with a base to form a salt, such as an ammonium salt; a base, or by other predetermined methods. Metal salts, such as sodium or potassium salts; alkaline earth metal salts, such as calcium or magnesium salts; salts of organic bases, such as dicyclohexylamine; N-methyl-D-reduced glucosamine; ginseng (hydroxymethyl) Methylamine and its salts with amino acids such as arginine, lysine and the like.

「腫瘤」在應用於經診斷患有或疑似患有癌症之個體時係指任何尺寸之惡性或潛在惡性贅瘤或組織塊狀物,且包括原發性腫瘤及繼發性贅瘤。實體腫瘤為通常不含有囊腫或液體區域之組織之異常生長或塊狀物。實體腫瘤之實例為肉瘤、癌瘤及淋巴瘤。白血病(血液癌)通常不形成實體腫瘤(國家癌症學會(National Cancer Institute),癌症術語詞典(Dictionary of Cancer Terms))。"Neoplasm" when applied to an individual diagnosed with or suspected of having cancer means a malignant or potentially malignant neoplasm or mass of tissue of any size and includes primary tumors and secondary neoplasms. Solid tumors are abnormal growths or masses of tissue that usually do not contain cysts or areas of fluid. Examples of solid tumors are sarcomas, carcinomas and lymphomas. Leukemias (blood cancers) do not usually form solid tumors (National Cancer Institute, Dictionary of Cancer Terms).

「腫瘤負荷(Tumor burden)」亦稱為「腫瘤負載」,係指分佈在體內之腫瘤材料的總量。腫瘤負荷係指整個身體,包括淋巴結及骨髓中癌細胞之總數或腫瘤之總尺寸。腫瘤負荷可藉由此項技術中已知之多種方法測定,諸如使用測徑器,或當在身體中時使用成像技術,例如超音波、骨骼掃描、電腦斷層攝影術(CT)或磁共振成像(MRI)掃描。"Tumor burden", also known as "tumor burden", refers to the total amount of tumor material distributed in the body. Tumor burden refers to the total number of cancer cells or the total size of the tumor in the entire body, including lymph nodes and bone marrow. Tumor burden can be determined by a variety of methods known in the art, such as using calipers, or when in the body using imaging techniques such as ultrasound, bone scans, computed tomography (CT) or magnetic resonance imaging ( MRI) scan.

術語「腫瘤尺寸(tumor size)」係指可以腫瘤之長度及寬度量測的腫瘤之總尺寸。腫瘤尺寸可藉由此項技術中已知之多種方法測定,諸如藉由自個體移出之後例如使用測徑器,或當在身體內時使用成像技術,例如骨骼掃描、超音波、CR或MRI掃描來量測腫瘤之尺寸。The term "tumor size" refers to the total size of the tumor as measured by the length and width of the tumor. Tumor size can be determined by a variety of methods known in the art, such as after removal from the subject, such as using a caliper, or while in the body using imaging techniques such as bone scans, ultrasound, CR or MRI scans. Measure the size of the tumor.

本發明係關於一種選擇患有具有同源重組修復缺陷之癌症之個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療之方法,其包含:a)根據癌症之活檢判定gLOH分數;及b)基於gLOH分數選擇個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療。該方法進一步包含向所選個體投與他拉唑帕尼或其醫藥學上可接受之鹽。The present invention relates to a method of selecting individuals with cancer having homologous recombination repair deficiency for treatment with talazopanib or a pharmaceutically acceptable salt thereof, comprising: a) determining a gLOH score based on a biopsy of the cancer ; and b) selecting individuals for treatment with talazopanib or a pharmaceutically acceptable salt thereof based on gLOH score. The method further includes administering talazopanib or a pharmaceutically acceptable salt thereof to the selected individual.

本發明係關於一種選擇患有具有同源重組修復缺陷之癌症之個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療之方法,包含:a)根據癌症之活檢判定gLOH分數;及b)基於該gLOH分數選擇個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療;且進一步包含向所選個體投與他拉唑帕尼或其醫藥學上可接受之鹽。The present invention relates to a method of selecting individuals suffering from cancer with homologous recombination repair deficiency for treatment with talazopanib or a pharmaceutically acceptable salt thereof, comprising: a) determining the gLOH score based on a biopsy of the cancer; and b) selecting an individual for treatment with talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score; and further comprising administering talazopanib or a pharmaceutically acceptable salt thereof to the selected individual .

本發明之方法適用於選擇個體以用他拉唑帕尼治療。特定言之,本發明之方法適用於選擇患有具有同源重組修復缺陷之癌症之個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療。The methods of the invention are suitable for selecting individuals for treatment with talazopanib. In particular, the methods of the present invention are suitable for selecting individuals with cancers having homologous recombination repair defects for treatment with talazopanib or a pharmaceutically acceptable salt thereof.

本發明係關於一種治療個體內具有同源重組修復缺陷之癌症之方法,其包含:a)根據癌症之活檢判定gLOH分數;及b)基於gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。The present invention relates to a method for treating cancer with homologous recombination repair deficiency in an individual, which includes: a) determining a gLOH score based on a biopsy of the cancer; and b) administering a therapeutically effective amount of talazopanib or talazopanib based on the gLOH score. Its pharmaceutically acceptable salt.

在一態樣中,本發明係關於一種他拉唑帕尼或其醫藥學上可接受之鹽在治療個體內之具有同源重組修復缺陷之癌症的用途,其包含:a)根據癌症之活檢判定gLOH分數;及b)基於gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。In one aspect, the invention relates to the use of talazopanib or a pharmaceutically acceptable salt thereof in the treatment of cancer with homologous recombination repair deficiency in an individual, comprising: a) based on a biopsy of the cancer Determine the gLOH score; and b) administer a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score.

在一態樣中,本發明係關於一種他拉唑帕尼或其醫藥學上可接受之鹽作為用於治療個體內之具有同源重組修復缺陷之癌症的藥劑的用途,其包含:a)根據癌症之活檢判定gLOH分數;及b)基於gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。In one aspect, the present invention relates to the use of talazopanib or a pharmaceutically acceptable salt thereof as an agent for treating cancer with homologous recombination repair deficiency in an individual, comprising: a) Determine the gLOH score based on the biopsy of the cancer; and b) administer a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score.

本發明之方法適用於治療癌症。特定言之,本發明之方法適用於治療具有同源重組修復缺陷之癌症。此外,本發明之方法適用於鑑別具有同源重組修復缺陷之癌症,該癌症對癌症治療敏感,諸如以他拉唑帕尼治療。在一些實施例中,所提供之方法導致以下作用之一或多者:(1)抑制癌細胞增殖;(2)抑制癌細胞侵襲性;(3)誘導癌細胞之細胞凋亡;(4)抑制癌細胞轉移;(5)抑制血管新生;或(6)克服與癌症治療相關之一或多種抗性機制。The method of the invention is suitable for treating cancer. In particular, the methods of the present invention are suitable for the treatment of cancers with defects in homologous recombination repair. Furthermore, the methods of the present invention are suitable for identifying cancers with homologous recombination repair defects that are sensitive to cancer treatments, such as treatment with talazopanib. In some embodiments, the provided methods result in one or more of the following effects: (1) inhibiting cancer cell proliferation; (2) inhibiting cancer cell invasiveness; (3) inducing apoptosis of cancer cells; (4) Inhibit cancer cell metastasis; (5) inhibit angiogenesis; or (6) overcome one or more resistance mechanisms related to cancer treatment.

在一個實施例中,本發明係關於一種鑑別對以他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,其包含:a)根據癌症之活檢判定基因雜合性缺失(gLOH)分數;及b)基於gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。In one embodiment, the present invention relates to a method for identifying cancers with homologous recombination repair defects that are sensitive to treatment with talazopanib or a pharmaceutically acceptable salt thereof, which comprises: a) based on the characteristics of the cancer Biopsy determines the gene loss of heterozygosity (gLOH) score; and b) administers a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score.

「基因雜合性缺失(Genomic loss of heterozygosity)」或「gLOH」係由同源重組修復(HRR)缺陷引起且產生基因以同型接合或半接合狀態存在之基因體區域。根據本發明之方法,基於gLOH分數選擇患有具有同源重組修復缺陷之癌症之個體以用他拉唑帕尼治療。gLOH分數可根據Sokol等人, JCO Precis Oncol. 2020;4:442-465判定,其提供gLOH量測及性能之詳細描述,具體如下:方法:使用FoundationOne ®CDx (Foundation Medicine,Inc.)檢定中定序的>3,500個多晶型單核苷酸多型性(SNP)的全基因體非整倍體/複本數圖譜和次要對偶基因頻率(AF),推斷出22條常染色體之LOH片段。使用比較基因體雜交類方法,藉由針對與處理匹配之正常對照標準化在所有外顯子及全基因體SNP獲得之序列涵蓋範圍,獲得樣品之對數比圖譜(Frampton GM等人: Nat. Biotechnol31:1023-1031, 2013)。使用經定序SNP之AF對此圖譜進行分段及解讀以估算各片段(i)處之複本數(Ci)及次要對偶基因計數(Mi)。若Ci≠0且Mi=0,則判定片段具有LOH。低腫瘤含量或低非整倍體係未能通過品質控制來進行gLOH推斷之最常見原因。gLOH百分比之計算將兩種類型之LOH片段排除在外:跨越整個染色體或染色體臂之≥90%之LOH片段因為此等LOH事件通常經由非HRD機制(例如,有絲分裂不分離)產生,及LOH推斷不明確之區域。對於各腫瘤,gLOH百分比計算為100×未排除之LOH區域之總長度(xi)除以基因體之未排除區域之總長度。 "Genomic loss of heterozygosity" or "gLOH" is caused by homologous recombination repair (HRR) defects and results in a region of the gene body where genes exist in a homozygous or semi-zygous state. According to the methods of the invention, individuals with cancers having homologous recombination repair defects are selected for treatment with talazopanib based on gLOH scores. The gLOH score can be determined according to Sokol et al., JCO Precis Oncol . 2020;4:442-465, which provides a detailed description of gLOH measurement and performance as follows: Method: Assayed using FoundationOne ® CDx (Foundation Medicine, Inc.) Sequenced genome-wide aneuploidy/copy number map and minor allele frequency (AF) of >3,500 polymorphic single nucleotide polymorphisms (SNPs), inferred LOH segments of 22 autosomal chromosomes . Using a comparative genomic hybridization-type approach, a log-ratio profile of the sample was obtained by normalizing the sequence coverage obtained at all exons and genome-wide SNPs against a normal control matched to the treatment (Frampton GM et al.: Nat. Biotechnol 31 :1023-1031, 2013). This map was segmented and interpreted using AF of sequenced SNPs to estimate the number of copies (Ci) and minor allele count (Mi) at each segment (i). If Ci≠0 and Mi=0, the segment is determined to have LOH. Low tumor content or low aneuploidy systems are the most common reasons for failure to pass quality control for gLOH inference. Calculation of percent gLOH excludes two types of LOH segments: LOH segments spanning ≥90% of the entire chromosome or chromosome arm because these LOH events typically arise via non-HRD mechanisms (e.g., mitotic nondisjunction), and LOH inference is not clear area. For each tumor, the percent gLOH was calculated as 100×the total length of the non-excluded LOH regions (xi) divided by the total length of the non-excluded regions of the genome.

在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約8.8%;至少約9%;至少約9.2%;至少約10%;至少約11%;至少約12%;至少約13%;至少約14%;至少約15%;至少約16%;至少約17%;至少約18%;至少約19%;至少約20%;至少約21%;至少約22%;至少約23%;至少約24%;或至少約25%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約24%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約23%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約22%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約21%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約20%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約19%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約18%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約17%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約16%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約15%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約14%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約13%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約12%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約11%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約10%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約9%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約8.8%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約8.3%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少約8.0%。In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 8.8%; at least about 9%; at least about 9.2%; at least about 10%; at least about 11%; at least about 12%; at least about 13%; At least about 14%; at least about 15%; at least about 16%; at least about 17%; at least about 18%; at least about 19%; at least about 20%; at least about 21%; at least about 22%; at least about 23%; At least about 24%; or at least about 25%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 24%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 23%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 22%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 21%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 20%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 19%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 18%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 17%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 16%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 15%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 14%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 13%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 12%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 11%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 10%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 9%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 8.8%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 8.3%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least about 8.0%.

在本發明之一個實施例中,其中gLOH分數或gLOH百分比係至少8.8%;至少9%;至少9.2%;至少10%;至少11%;至少12%;至少13%;至少14%;至少15%;至少16%;至少17%;至少18%;至少19%;至少20%;至少21%;至少22%;至少23%;至少24%;或至少25%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少24%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少23%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少22%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少21%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少20%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少19%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少18%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少17%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少16%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少15%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少14%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少13%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少12%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少11%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少10%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少9%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少8.8%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少8.3%。在本發明之一個實施例中,gLOH分數或gLOH百分比係至少8.0%。In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 8.8%; at least 9%; at least 9.2%; at least 10%; at least 11%; at least 12%; at least 13%; at least 14%; at least 15 %; at least 16%; at least 17%; at least 18%; at least 19%; at least 20%; at least 21%; at least 22%; at least 23%; at least 24%; or at least 25%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 24%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 23%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 22%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 21%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 20%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 19%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 18%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 17%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 16%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 15%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 14%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 13%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 12%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 11%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 10%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 9%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 8.8%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 8.3%. In one embodiment of the invention, the gLOH fraction or gLOH percentage is at least 8.0%.

下一代定序(NGS)係使用大規模平行處理之概念進行DNA定序之若干高通量方法之任一者。雖然存在使用不同定序技術之許多不同NGS平台,但所有NGS平台並行執行數百萬個DNA小片段之定序。熟習此項技術者熟悉下一代定序。Next generation sequencing (NGS) is any of several high-throughput methods for DNA sequencing that use the concept of massively parallel processing. Although there are many different NGS platforms using different sequencing technologies, all NGS platforms perform sequencing of millions of small DNA fragments in parallel. Those skilled in the art are familiar with next generation sequencing.

根據本發明之方法,癌症或腫瘤中之同源重組修復缺陷可使用下一代定序判定。According to the methods of the present invention, homologous recombination repair defects in cancer or tumors can be determined using next generation sequencing.

根據本發明之方法,根據癌症之活檢的gLOH分數可使用下一代定序判定。例如,可使用能夠評定gLOH之基於小組之定序分析,諸如FoundationOne®CDx (Foundation Medicine, Inc.)。gLOH可用作精選婦科疾病中之FoundationOne®CDx測試之一部分。已報導DDR/HRR基因中之雙對偶基因改變與一系列腫瘤類型之增加之gLOH相關聯,包括可得益於PARP抑制劑之彼等腫瘤類型,諸如乳癌、卵巢癌、前列腺癌及胰臟癌(Westphalen B等人, Clin Cancer Res.2021)。然而,一般而言,gLOH分數在前列腺癌中相對較低且並不展現在乳癌及卵巢癌中看到之動態範圍(Sokol ES等人. JCO Precis Oncol.2020;4:442-465)。 According to the methods of the present invention, next generation sequencing can be used to determine the gLOH score from a cancer biopsy. For example, a panel-based sequencing assay capable of assessing gLOH can be used, such as FoundationOne® CDx (Foundation Medicine, Inc.). gLOH is available as part of the FoundationOne® CDx test in select gynecological conditions. Dual genetic alterations in DDR/HRR genes have been reported to be associated with increased gLOH in a range of tumor types, including those that benefit from PARP inhibitors, such as breast, ovarian, prostate, and pancreatic cancers (Westphalen B et al., Clin Cancer Res. 2021). However, in general, gLOH scores are relatively low in prostate cancer and do not exhibit the dynamic range seen in breast and ovarian cancer (Sokol ES et al. JCO Precis Oncol. 2020;4:442-465).

一種選擇患有具有同源重組修復缺陷之癌症之個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療之方法,其包含:a)根據癌症之活檢判定基因雜合性缺失(gLOH)分數;及b)若gLOH分數係至少8.8%,則選擇個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療。A method of selecting individuals with cancer having homologous recombination repair deficiency for treatment with talazopanib or a pharmaceutically acceptable salt thereof, comprising: a) determining loss of gene heterozygosity based on a biopsy of the cancer ( gLOH) score; and b) if the gLOH score is at least 8.8%, select the individual for treatment with talazopanib or a pharmaceutically acceptable salt thereof.

一種鑑別對以他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,其包含:a)根據癌症之活檢判定基因雜合性缺失(gLOH)分數;及b)若gLOH分數係至少8.8%,則投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。A method of identifying cancers with homologous recombination repair defects that are susceptible to treatment with talazopanib or a pharmaceutically acceptable salt thereof, comprising: a) determining genetic loss of heterozygosity (gLOH) based on a biopsy of the cancer score; and b) if the gLOH score is at least 8.8%, administer a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof.

在一個實施例中,本發明係關於一種治療個體內具有同源重組修復缺陷之癌症之方法,其包含:a)根據癌症之活檢判定gLOH分數;及b)若gLOH分數係至少8.8%,則投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。In one embodiment, the invention relates to a method of treating cancer in an individual with homologous recombination repair deficiency, comprising: a) determining the gLOH score based on a biopsy of the cancer; and b) if the gLOH score is at least 8.8%, then Administer a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof.

在另一態樣中,本發明係關於一種他拉唑帕尼或其醫藥學上可接受之鹽在治療個體中具有同源重組修復缺陷之癌症的用途,其包含:a)根據癌症之活檢判定gLOH分數;及b)若gLOH分數係至少8.8%,則投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。In another aspect, the invention relates to the use of talazopanib or a pharmaceutically acceptable salt thereof in the treatment of cancer in an individual with homologous recombination repair deficiency, comprising: a) based on a biopsy of the cancer Determine the gLOH score; and b) if the gLOH score is at least 8.8%, administer a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof.

在另一態樣中,本發明係關於一種他拉唑帕尼或其醫藥學上可接受之鹽作為藥劑用於治療個體中具有同源重組修復缺陷之癌症的用途,其包含:a)根據癌症之活檢判定gLOH分數;及b)若gLOH分數係至少8.8%,則投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。In another aspect, the present invention relates to the use of talazopanib or a pharmaceutically acceptable salt thereof as a medicament for the treatment of cancer in an individual with homologous recombination repair deficiency, comprising: a) according to Biopsy of the cancer determines the gLOH score; and b) if the gLOH score is at least 8.8%, administer a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof.

在本發明之一個實施例中,個體係哺乳動物。In one embodiment of the invention, the individual is a mammal.

在本發明之一個實施例中,個體係人類。In one embodiment of the invention, the individual system is human.

在實施例中,本發明之方法可能適用於治療癌症,包括但不限於以下之癌症: 循環系統,例如心臟(肉瘤[血管肉瘤、纖維肉瘤、橫紋肌肉瘤、脂肪肉瘤]、黏液瘤、橫紋肌瘤、纖維瘤、脂肪瘤及畸胎瘤)、縱隔及胸膜,及其他胸內器官、血管腫瘤及腫瘤相關血管組織; 呼吸道,例如鼻腔及中耳、副鼻竇、喉、氣管、支氣管及肺,諸如小細胞肺癌(SCLC)、非小細胞肺癌(NSCLC)、支氣管癌瘤(鱗狀細胞、未分化小細胞、未分化大細胞、腺癌)、肺泡(支氣管)癌瘤、支氣管腺瘤、肉瘤、淋巴瘤、軟骨瘤性錯構瘤、間皮瘤; 胃腸道系統,例如食道(鱗狀細胞癌瘤、腺癌、平滑肌肉瘤、淋巴瘤)、胃部(癌瘤、淋巴瘤、平滑肌肉瘤)、胃、胰臟(導管腺癌、胰島素瘤、升糖素瘤、胃泌素瘤、類癌、血管活性腸肽瘤)、小腸(腺癌、淋巴瘤、類癌、卡波西氏肉瘤(Karposi's sarcoma)、平滑肌瘤、血管瘤、脂肪瘤、神經纖維瘤、纖維瘤)、大腸(腺癌、導管腺瘤、纖毛狀腺瘤、錯構瘤、平滑肌瘤); 泌尿生殖道,例如腎臟(腺癌、威爾姆氏腫瘤(Wilm's tumor)[腎母細胞瘤]、淋巴瘤、白血病)、膀胱及/或尿道(鱗狀細胞癌、移行細胞或尿道上皮癌瘤、腺癌)、前列腺(腺癌、肉瘤)、睾丸(精原細胞瘤、畸胎瘤、胚胎性癌、畸胎癌、絨毛膜癌瘤、肉瘤、間質細胞癌瘤、纖維瘤、纖維腺瘤、腺瘤樣腫瘤、脂肪瘤); 肝臟(例如肝癌、肝細胞癌瘤)、膽管癌、肝母細胞瘤、血管肉瘤、肝細胞腺瘤、血球瘤、胰臟內分泌腫瘤(諸如嗜鉻細胞瘤、胰島素瘤、血管活性腸道肽腫瘤、胰島細胞腫瘤及升糖素瘤); 骨骼,例如成骨性肉瘤(骨肉瘤)、纖維肉瘤、惡性纖維組織細胞瘤、軟骨肉瘤、尤文氏肉瘤(Ewing's sarcoma)、惡性淋巴瘤(網狀細胞肉瘤)、多發性骨髓瘤、惡性巨細胞瘤脊索瘤、骨軟骨瘤(骨軟骨外生骨疣)、良性軟骨瘤、軟骨母細胞瘤、軟骨黏液性纖維瘤、骨樣骨瘤及巨細胞瘤; 神經系統,例如中樞神經系統(CNS)之贅瘤、原發性CNS淋巴瘤、頭骨癌(骨瘤、血管瘤、肉芽腫瘤、黃瘤、骨炎畸形)、腦膜(脊膜瘤、脊膜肉瘤、神經膠質瘤)、腦癌(星形細胞瘤、神經管胚細胞瘤、神經膠瘤、室管膜瘤、胚細胞瘤[松果體瘤]、多形性神經膠母細胞瘤、少突神經膠質瘤、神經鞘瘤、視網膜母細胞瘤、先天性腫瘤)、脊髓神經纖維瘤、脊膜瘤、神經膠瘤、肉瘤; 生殖系統,例如婦科、子宮(子宮內膜癌)、子宮頸(子宮頸癌、腫瘤前子宮頸發育不良)、卵巢(卵巢癌[漿液性囊腺癌、黏液性囊腺癌、未分類之癌瘤]、濾泡膜細胞瘤、塞特利氏-萊迪希氏細胞腫瘤(Sertoli-Leydig cell tumor)、無性細胞瘤、惡性畸胎瘤)、外陰(鱗狀細胞癌、上皮內癌瘤、腺癌、纖維肉瘤、黑色素瘤)、陰道(透明細胞癌瘤、鱗狀細胞癌、葡萄樣肉瘤(胚胎性橫紋肌肉瘤))、輸卵管(癌瘤)及與女性生殖器官相關聯之其他部位;胎盤、陰莖、前列腺、睾丸及與男性生殖器官相關聯之其他部位; 血液系統,例如血液(骨髓白血病[急性及慢性]、急性淋巴母細胞白血病、慢性淋巴球性白血病、骨髓增生病、多發性骨髓瘤、骨髓發育不良症候群)、霍奇金氏疾病(Hodgkin's disease)、非霍奇金氏淋巴瘤[惡性淋巴瘤]; 口腔,例如唇、舌片、齒齦、口底、齶及口部之其他部分、腮腺、及唾液腺之其他部分、扁桃體、口咽、鼻咽、梨狀窩、喉咽、及唇部之其他部位、口腔及咽; 皮膚,例如惡性黑色素瘤、皮膚黑色素瘤、基底細胞癌、鱗狀細胞癌、卡波西氏肉瘤、發育不良痣、脂肪瘤、血管瘤、皮膚纖維瘤及瘢痕瘤; 腎上腺:神經母細胞瘤;及 其他組織,包括結締組織及軟組織、腹膜後腔及腹膜、眼睛、眼內黑色素瘤及附件、乳房、頭部或/及頸部、肛門區、甲狀腺、副甲狀腺、腎上腺及其他內分泌腺體及相關結構、淋巴結之繼發性及未指明的惡性贅瘤、呼吸及消化系統之繼發性惡性贅瘤及其他部位之繼發性惡性贅瘤。 In embodiments, the methods of the present invention may be suitable for treating cancer, including but not limited to the following cancers: Circulatory system, such as heart (sarcomas [angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma], myxoma, rhabdomyosarcoma, fibroma, lipoma and teratoma), mediastinum and pleura, and other intrathoracic organs, vascular tumors and tumor-related vascular tissue; Respiratory tract, such as nasal cavity and middle ear, paranasal sinuses, larynx, trachea, bronchi and lungs, such as small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), bronchial carcinoma (squamous cell, undifferentiated small cell, undifferentiated Large cell, adenocarcinoma), alveolar (bronchial) carcinoma, bronchial adenoma, sarcoma, lymphoma, enchondromatous hamartoma, mesothelioma; Gastrointestinal system, such as esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), stomach, pancreas (ductal adenocarcinoma, insulinoma, glycemic cystoma, gastrinoma, carcinoid, vasoactive intestinal peptide tumor), small intestine (adenocarcinoma, lymphoma, carcinoid, Karposi's sarcoma (Karposi's sarcoma), leiomyoma, hemangioma, lipoma, nerve Fibroids, fibroids), large intestine (adenocarcinoma, ductal adenoma, ciliated adenoma, hamartoma, leiomyoma); Genitourinary tract, such as kidney (adenocarcinoma, Wilm's tumor [Wilm's tumor], lymphoma, leukemia), bladder and/or urethra (squamous cell carcinoma, transitional cell or urothelial carcinoma) , adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratoma, choriocarcinoma, sarcoma, stromal cell carcinoma, fibroma, fibroglandular tumors, adenomatoid tumors, lipomas); Liver (such as liver cancer, hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hematoma, pancreatic endocrine tumors (such as pheochromocytoma, insulinoma, vasoactive intestinal peptide tumors , islet cell tumors and glucagonoma); Bones, such as osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell Chordoma, osteochondroma (osteochondral exostosis), benign enchondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumor; Nervous system, such as tumors of the central nervous system (CNS), primary CNS lymphoma, skull cancer (osteoma, hemangioma, granuloma, xanthoma, osteitis malformation), meninges (meningioma, meningiosarcoma) , glioma), brain cancer (astrocytoma, medulloblastoma, glioma, ependymoma, blastoma [pineal tumor], glioblastoma multiforme, oligodendroma Glioma, schwannoma, retinoblastoma, congenital tumors), spinal neurofibroma, meningioma, glioma, sarcoma; Reproductive system, such as gynecology, uterus (endometrial cancer), cervix (cervical cancer, preneoplastic cervical dysplasia), ovary (ovarian cancer [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma] tumor], theca cell tumor, Sertoli-Leydig cell tumor (Sertoli-Leydig cell tumor), dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma) , adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonic rhabdomyosarcoma)), fallopian tube (carcinoma) and other parts associated with female reproductive organs; Placenta, penis, prostate, testicles and other parts associated with male reproductive organs; Blood system, such as blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndromes), Hodgkin's disease , non-Hodgkin's lymphoma [malignant lymphoma]; Oral cavity, such as lips, tongue, gingiva, floor of the mouth, palate and other parts of the mouth, parotid glands and other parts of the salivary glands, tonsils, oropharynx, nasopharynx, piriform fossa, hypopharynx, and other parts of the lips , oral cavity and pharynx; Skin, such as malignant melanoma, cutaneous melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, dysplastic nevus, lipoma, hemangioma, dermatofibroma and keloid; Adrenal gland: neuroblastoma; and Other tissues, including connective and soft tissue, retroperitoneal cavity and peritoneum, eye, intraocular melanoma and adnexa, breast, head or/and neck, anal area, thyroid, parathyroid, adrenal and other endocrine glands and related Secondary and unspecified malignant neoplasms of structures, lymph nodes, secondary malignant neoplasms of respiratory and digestive systems, and secondary malignant neoplasms of other parts.

當在本文中結合本發明使用時,「癌症」之實例包括選自以下之癌症:肺癌(NSCLC及SCLC)、乳癌(包括三陰性乳癌、激素陽性乳癌、HER2陰性乳癌、HER2陽性乳癌及三陽性乳癌)、卵巢癌、大腸癌、直腸癌、肛門區癌、前列腺癌(包括去勢敏感性或激素敏感性前列腺癌及激素難治性前列腺癌,亦稱為去勢抗性前列腺癌)、肝細胞癌瘤、彌漫性大B細胞淋巴瘤、濾泡性淋巴瘤、黑色素瘤及唾液腺腫瘤,或前述癌症之一或多者的組合。When used herein in connection with the present invention, examples of "cancer" include cancers selected from the group consisting of: lung cancer (NSCLC and SCLC), breast cancer (including triple-negative breast cancer, hormone-positive breast cancer, HER2-negative breast cancer, HER2-positive breast cancer, and triple-positive breast cancer). Breast cancer), ovarian cancer, colorectal cancer, rectal cancer, anal area cancer, prostate cancer (including castration-sensitive or hormone-sensitive prostate cancer and hormone-refractory prostate cancer, also known as castration-resistant prostate cancer), hepatocellular carcinoma , diffuse large B-cell lymphoma, follicular lymphoma, melanoma and salivary gland tumors, or a combination of one or more of the foregoing cancers.

當在本文中結合本發明使用時,「癌症」之實例包括選自以下之癌症:肺癌(NSCLC及SCLC)、乳癌(包括三陰性乳癌、激素陽性乳癌及HER2陰性乳癌)、卵巢癌、前列腺癌(包括去勢敏感性或激素敏感性前列腺癌及激素難治性前列腺癌,亦稱為去勢抗性前列腺癌),或前述癌症之一或多者的組合。When used herein in connection with the present invention, examples of "cancer" include cancers selected from the group consisting of: lung cancer (NSCLC and SCLC), breast cancer (including triple-negative breast cancer, hormone-positive breast cancer and HER2-negative breast cancer), ovarian cancer, prostate cancer (including castration-sensitive or hormone-sensitive prostate cancer and hormone-refractory prostate cancer, also known as castration-resistant prostate cancer), or a combination of one or more of the foregoing cancers.

當在本文中結合本發明使用時,「癌症」之實例包括選自以下之癌症:前列腺癌、雄激素受體陽性乳癌、肝細胞癌瘤、及唾液腺腫瘤,或前述癌症之一或多者的組合。As used herein in connection with the present invention, examples of "cancer" include cancers selected from the group consisting of prostate cancer, androgen receptor-positive breast cancer, hepatocellular carcinoma, and salivary gland tumors, or one or more of the foregoing. combination.

當在本文中結合本發明使用時,「癌症」之實例包括選自以下之癌症:雄激素受體陽性乳癌、肝細胞癌瘤、及唾液腺腫瘤,或前述癌症之一或多者的組合。As used herein in connection with the present invention, examples of "cancer" include cancers selected from the group consisting of androgen receptor-positive breast cancer, hepatocellular carcinoma, and salivary gland tumors, or a combination of one or more of the foregoing.

當在本文中結合本發明使用時,「癌症」之實例包括選自以下之癌症:三陰性乳癌、激素陽性乳癌、HER2陰性乳癌、三陽性乳癌、去勢敏感性前列腺癌、去勢抗性前列腺癌、肝細胞癌瘤、及唾液腺腫瘤或前述癌症之一或多者的組合。When used herein in connection with the present invention, examples of "cancer" include cancers selected from the group consisting of triple-negative breast cancer, hormone-positive breast cancer, HER2-negative breast cancer, triple-positive breast cancer, castration-sensitive prostate cancer, castration-resistant prostate cancer, Hepatocellular carcinoma, salivary gland tumors, or a combination of one or more of the foregoing cancers.

當在本文中結合本發明使用時,「癌症」之實例包括選自以下之癌症:三陰性乳癌、激素陽性乳癌、及HER2陰性乳癌,或前述癌症之一或多者的組合。As used herein in connection with the present invention, examples of "cancer" include cancers selected from the group consisting of triple-negative breast cancer, hormone-positive breast cancer, and HER2-negative breast cancer, or a combination of one or more of the foregoing.

當在本文中結合本發明使用時,「癌症」之實例包括選自以下之癌症:去勢敏感性前列腺癌及去勢抗性前列腺癌,或前述癌症之一或多者的組合。As used herein in connection with the present invention, examples of "cancer" include cancers selected from the group consisting of castration-sensitive prostate cancer and castration-resistant prostate cancer, or a combination of one or more of the foregoing.

在本發明之一個實施例中,癌症係實體腫瘤。In one embodiment of the invention, the cancer is a solid tumor.

在本發明之一個實施例中,癌症係實體腫瘤,該實體腫瘤係雄激素依賴性的。In one embodiment of the invention, the cancer is a solid tumor that is androgen dependent.

在本發明之一個實施例中,癌症係實體腫瘤,該實體腫瘤表現雄激素受體。In one embodiment of the invention, the cancer is a solid tumor that expresses androgen receptors.

在一個實施例中,癌症係前列腺癌。In one embodiment, the cancer is prostate cancer.

在一個實施例中,癌症係高風險前列腺癌。In one embodiment, the cancer is high risk prostate cancer.

在一個實施例中,癌症係局部晚期前列腺癌。In one embodiment, the cancer is locally advanced prostate cancer.

在一個實施例中,癌症係高風險局部晚期前列腺癌。In one embodiment, the cancer is high risk locally advanced prostate cancer.

在一個實施例中,癌症係轉移性前列腺癌。In one embodiment, the cancer is metastatic prostate cancer.

在一個實施例中,癌症係激素敏感性前列腺癌,亦稱為去勢敏感性前列腺癌。激素敏感性前列腺癌通常特徵為組織學或細胞學上經確證之前列腺之腺癌,其仍對雄激素去除療法有反應。In one embodiment, the cancer is hormone-sensitive prostate cancer, also known as castration-sensitive prostate cancer. Hormone-sensitive prostate cancer is usually characterized by histologically or cytologically confirmed adenocarcinoma of the prostate that remains responsive to androgen ablation therapy.

在一個實施例中,癌症係非轉移性激素敏感性前列腺癌。In one embodiment, the cancer is non-metastatic hormone-sensitive prostate cancer.

在一個實施例中,癌症係高風險非轉移性激素敏感性前列腺癌。In one embodiment, the cancer is high-risk non-metastatic hormone-sensitive prostate cancer.

在一個實施例中,癌症係轉移性激素敏感性前列腺癌。In one embodiment, the cancer is metastatic hormone-sensitive prostate cancer.

在一個實施例中,癌症係去勢敏感性前列腺癌。In one embodiment, the cancer is castration-sensitive prostate cancer.

在一個實施例中,癌症係非轉移性去勢敏感性前列腺癌。In one embodiment, the cancer is non-metastatic castration-sensitive prostate cancer.

在一個實施例中,癌症係轉移性去勢敏感性前列腺癌。In one embodiment, the cancer is metastatic castration-sensitive prostate cancer.

在一個實施例中,癌症係具有DDR突變之去勢敏感性前列腺癌。在一個實施例中,癌症係具有DDR突變之非轉移性去勢敏感性前列腺癌。在一個實施例中,癌症係具有DDR突變之轉移性去勢敏感性前列腺癌。突變之DDR基因包括但不限於ATM、ATR、BRCA1、BRCA2、CDK12、CHEK2、FANCA、MLH1、MRE11A、NBN、PALB2及RAD51C。In one embodiment, the cancer is castration-sensitive prostate cancer harboring a DDR mutation. In one embodiment, the cancer is non-metastatic castration-sensitive prostate cancer harboring a DDR mutation. In one embodiment, the cancer is metastatic castration-sensitive prostate cancer harboring a DDR mutation. Mutated DDR genes include, but are not limited to, ATM, ATR, BRCA1, BRCA2, CDK12, CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2 and RAD51C.

在一個實施例中,癌症係去勢抗性前列腺癌,亦稱為激素難治性前列腺癌或雄激素非依賴性前列腺癌。去勢抗性前列腺癌通常特徵為在睾固酮之去勢含量(例如≤1.7 nmol/L睾固酮之含量或≤50 ng/dL睾固酮之含量)之設定下,組織學或細胞學上確證之前列腺之腺癌為去勢抗性的(例如,定義為PSA之2次或更多次連續升高,各評估之間的間隔≥1週,視情況導致最低點上方之2次或更多次50%或更高之增加,PSA含量≥2 ng/mL),睾固酮之去勢含量藉由雄激素去除療法及/或在睾丸切除術後實現。In one embodiment, the cancer is castration-resistant prostate cancer, also known as hormone-refractory prostate cancer or androgen-independent prostate cancer. Castration-resistant prostate cancer is typically characterized by histological or cytological confirmation in the setting of castrate levels of testosterone (e.g., ≤1.7 nmol/L testosterone or ≤50 ng/dL testosterone). The adenocarcinoma of the prostate is castration-resistant (e.g., defined as 2 or more consecutive increases in PSA, with ≥1 week between assessments, leading to 2 or more increases above the nadir as appropriate) A 50% or greater increase in PSA levels ≥2 ng/mL), castration of testosterone is achieved by androgen ablation therapy and/or after orchiectomy.

在一個實施例中,癌症係去勢抗性前列腺癌。In one embodiment, the cancer is castration-resistant prostate cancer.

在一個實施例中,癌症係非轉移性去勢抗性前列腺癌。In one embodiment, the cancer is non-metastatic castration-resistant prostate cancer.

在一個實施例中,癌症係轉移性去勢抗性前列腺癌。In one embodiment, the cancer is metastatic castration-resistant prostate cancer.

在一個實施例中,癌症係具有突變之去勢抗性前列腺癌。在一個實施例中,癌症係具有DDR突變之非轉移性去勢抗性前列腺癌。在一個實施例中,癌症係具有DDR突變之轉移性去勢抗性前列腺癌。突變之DDR基因包括但不限於ATM、ATR、BRCA1、BRCA2、CDK12、CHEK2、FANCA、MLH1、MRE11A、NBN、PALB2及RAD51C。In one embodiment, the cancer is castration-resistant prostate cancer harboring a mutation. In one embodiment, the cancer is non-metastatic castration-resistant prostate cancer harboring a DDR mutation. In one embodiment, the cancer is metastatic castration-resistant prostate cancer harboring a DDR mutation. Mutated DDR genes include, but are not limited to, ATM, ATR, BRCA1, BRCA2, CDK12, CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2 and RAD51C.

在一個實施例中,癌症係乳癌。In one embodiment, the cancer is breast cancer.

在一個實施例中,癌症係局部晚期或轉移性乳癌。In one embodiment, the cancer is locally advanced or metastatic breast cancer.

在一個實施例中,癌症係三陰性乳癌。In one embodiment, the cancer is triple negative breast cancer.

在一個實施例中,癌症係激素陽性乳癌,包括雌激素陽性乳癌及/或孕酮陽性乳癌。In one embodiment, the cancer is hormone-positive breast cancer, including estrogen-positive breast cancer and/or progesterone-positive breast cancer.

在一個實施例中,癌症係HER2陰性乳癌。In one embodiment, the cancer is HER2 negative breast cancer.

在一個實施例中,癌症係生殖系BRCA突變之HER2陰性乳癌。In one embodiment, the cancer is germline BRCA-mutated HER2-negative breast cancer.

在一個實施例中,癌症係HER2陽性乳癌。In one embodiment, the cancer is HER2-positive breast cancer.

在一個實施例中,癌症係三陽性乳癌。In one embodiment, the cancer is triple positive breast cancer.

在一個實施例中,癌症係卵巢癌。In one embodiment, the cancer is ovarian cancer.

在一個實施例中,癌症係小細胞肺癌。In one embodiment, the cancer is small cell lung cancer.

在一個實施例中,癌症係尤文氏肉瘤。In one embodiment, the cancer is Ewing's sarcoma.

在一個實施例中,癌症係肝細胞癌瘤。In one embodiment, the cancer is hepatocellular carcinoma.

在一個實施例中,癌症係唾液腺腫瘤。In one embodiment, the cancer is a salivary gland tumor.

在一個實施例中,癌症係局部晚期。In one embodiment, the cancer is locally advanced.

在一個實施例中,癌症係非轉移性的。In one embodiment, the cancer is non-metastatic.

在一個實施例中,癌症係轉移性的。In one embodiment, the cancer is metastatic.

在一個實施例中,癌症係難治性的。In one embodiment, the cancer is refractory to treatment.

在一個實施例中,癌症復發。In one embodiment, the cancer recurs.

在一個實施例中,癌症不耐受標準治療。In one embodiment, the cancer is intolerant to standard treatments.

在本發明之一個實施例中,本方法向經診斷患有癌症之個體投與,該癌症已出現對治療之抗性。In one embodiment of the invention, the method is administered to an individual diagnosed with cancer that has become resistant to treatment.

在另一態樣中,本發明之方法可另外包含投與其他抗癌劑,諸如抗腫瘤劑、抗血管生成劑、信號轉導抑制劑及抗增殖劑,其量一起能有效治療該癌症。在一些此類實施例中,抗腫瘤劑係選自由以下組成之群:有絲分裂抑制劑、烷基化劑、抗代謝物、嵌入抗生素、生長因子抑制劑、放射劑、細胞週期抑制劑、酶類、拓樸異構酶抑制劑、生物反應調節劑、抗體、細胞毒性劑、抗激素劑、雄激素去除療法及抗雄激素劑。在本發明之一個實施例中,其他抗癌劑係抗雄激素劑。在本發明之一個實施例中,抗雄激素劑係恩雜魯胺或阿帕盧醯胺(apalutamide)。In another aspect, the methods of the present invention may additionally comprise administering other anti-cancer agents, such as anti-neoplastic agents, anti-angiogenic agents, signal transduction inhibitors, and anti-proliferative agents, in amounts that together are effective to treat the cancer. In some such embodiments, the anti-neoplastic agent is selected from the group consisting of: mitotic inhibitors, alkylating agents, antimetabolites, intercalating antibiotics, growth factor inhibitors, radioactive agents, cell cycle inhibitors, enzymes , topoisomerase inhibitors, biological response modifiers, antibodies, cytotoxic agents, antihormonal agents, androgen ablation therapy and anti-androgens. In one embodiment of the invention, the other anti-cancer agent is an anti-androgen agent. In one embodiment of the invention, the antiandrogen is enzalutamide or apalutamide.

實例Example 11 : 根據他拉唑帕尼單藥療法在具有Based on talazopanib monotherapy in patients with DNADNA 修復改變fix changes (TALAPRO-1)(TALAPRO-1) 之轉移性去勢抗性前列腺癌中之開放標記Open labeling in metastatic castration-resistant prostate cancer 22 期試驗的基因體分析Genomic analysis of phase 1 trials : gLOHgLOH 分數及關聯探索性相關分析Exploratory correlation analysis of scores and associations 方法method

臨床試驗設計:在患有具有DNA修復改變之轉移性去勢抗性前列腺癌之病患中進行PARP抑制劑他拉唑帕尼之開放標記II期臨床試驗(de Bono等人, Lancet Oncol.2021年9月;22(9):1250-1264)。 Clinical trial design: Open-label phase II clinical trial of the PARP inhibitor talazopanib in patients with metastatic castration-resistant prostate cancer with DNA repair alterations (de Bono et al., Lancet Oncol. 2021 Sep;22(9):1250-1264).

合格準則 符合條件的病患係患有腺癌組織學之漸進性轉移性去勢抗性前列腺癌的18歲或以上之男性。疾病進展定義為讀數之間具有至少1週之時間間隔的至少三個遞增前列腺特異性抗原(PSA)值,如實體腫瘤反應評估標準版本1.1(RECIST 1.1)所定義之軟組織疾病惡化,或藉由前列腺癌工作組3 (PCWG3)標準所定義在骨骼掃描上具有兩個或更多個新轉移性病灶的骨骼疾病進展。若僅僅藉由PSA進展限定候選者,則篩選中心實驗室PSA值需要為2 µg/L或更高。臨床試驗納入標準在開始之後經過修正。原始臨床試驗設計允許在兩個重疊群組中登記具有可量測及不可量測疾病的病患:群組A,其包括具有在直接或間接涉及HRR之基因中可能對PARP抑制敏感之DDR改變的病患;及群組B,其包括在擴展之基因體中具有很可能或可能對PARP抑制敏感之DNA缺陷的病患。在於2018年2月15日協定修正案三之批准之情況下,登記限於具有可量測疾病的病患及具有可能對PARP抑制敏感之DNA改變的病患,其最初包含一組13個基因。FANCD2及FANCI未通過後續驗證要求,留下以下一組之11個HRR基因用於分析:ATM、ATR、BRCA1、BRCA2、CHEK2、FANCA、MLH1、MRE11A、NBN、PALB2及RAD51C(被稱作「DDR11」)。其他納入標準係東部腫瘤合作組(ECOG)效能狀態為0-2;接受雙側睾丸切除術或使用促性腺激素釋放激素促效劑或拮抗劑之進行中之雄激素去除療法,篩選時血清睾固酮為50 ng/dL或更低(≤1.73 nmol/L);對於接受此等療法的病患,穩定雙膦酸鹽或地舒單抗劑量持續至少4週;估算之預期壽命為至少6個月(由研究人員估算);及先前在轉移性情況(去勢敏感性前列腺癌或去勢抗性前列腺癌;病患可能已接受鐳233或卡巴他賽(cabazitaxel),或兩者)下接受過一種或兩種化學治療方案(≥1種基於紫杉烷之)治療及針對轉移性去勢抗性前列腺癌進展使用至少一種新穎激素療法(恩雜魯胺、阿比特龍或兩者)。 Eligibility criteria : Eligible patients are men 18 years of age or older with progressive metastatic castration-resistant prostate cancer with adenocarcinoma histology. Disease progression was defined as at least three increasing prostate-specific antigen (PSA) values with a time interval of at least 1 week between readings, worsening of soft tissue disease as defined by Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), or by progression of soft tissue disease Progression of skeletal disease with two or more new metastatic lesions on bone scan as defined by Prostate Cancer Working Group 3 (PCWG3) criteria. To qualify candidates solely by PSA progression, the screening central laboratory PSA value needs to be 2 µg/L or higher. Clinical trial inclusion criteria were revised after initiation. The original clinical trial design allowed for the enrollment of patients with measurable and non-measurable disease in two overlapping cohorts: Cohort A, which included patients with DDR alterations that may be sensitive to PARP inhibition in genes directly or indirectly involved in HRR patients; and Group B, which includes patients with DNA defects in the expanded genome that are likely or likely to be sensitive to PARP inhibition. Subject to the approval of Amendment 3 of the Agreement on February 15, 2018, registration was limited to patients with measurable disease and patients with DNA alterations that may be sensitive to PARP inhibition, which initially comprised a panel of 13 genes. FANCD2 and FANCI failed subsequent validation requirements, leaving the following group of 11 HRR genes for analysis: ATM, ATR, BRCA1, BRCA2, CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2 and RAD51C (called "DDR11 ”). Other inclusion criteria were Eastern Cooperative Oncology Group (ECOG) performance status 0-2; undergoing bilateral orchiectomy or ongoing androgen ablation therapy using gonadotropin-releasing hormone agonists or antagonists, and serum testosterone levels at screening Ketones 50 ng/dL or less (≤1.73 nmol/L); stable bisphosphonate or denosumab dose for at least 4 weeks for patients receiving these therapies; estimated life expectancy is at least 6 months months (estimated by investigators); and previously received one in the setting of metastatic disease (castrate-sensitive prostate cancer or castration-resistant prostate cancer; patients may have received radium-233 or cabazitaxel, or both) or two chemotherapy regimens (≥1 taxane-based) and at least one novel hormonal therapy (enzalutamide, abiraterone, or both) for progression of metastatic castration-resistant prostate cancer.

治療 向病患每天經口給予他拉唑帕尼1 mg (或對於具有中度腎臟損傷之病患每天0.75 mg,定義為每1.73 m²,30-59 mL/min的經估算腎小球濾過率),針對自3級或4級不良事件恢復,給予劑量修正或適當支援性護理或兩者。持續投與他拉唑帕尼直至有進展,如根據放射照相成像、不可接受之毒性、研究人員決策、撤回同意書或死亡判定所判斷。單獨的PSA增加或循環腫瘤細胞計數增加並非中斷他拉唑帕尼之原因。 Treatment : Patients were administered talazopanib 1 mg orally daily (or 0.75 mg daily for patients with moderate renal impairment, defined as an estimated glomerular filtration of 30-59 mL/min per 1.73 m² rate), provide dose modification or appropriate supportive care, or both, for recovery from a Grade 3 or 4 adverse event. Administration of talazopanib was continued until progression, as judged by radiographic imaging, unacceptable toxicity, investigator decision-making, withdrawal of consent, or determination of death. An increase in PSA or circulating tumor cell count alone is not a reason to discontinue talazopanib.

評估腫瘤反應:在前24週期間每8週,然後在其後之每12週進行放射照相評估(腹部及骨盆之CT[較佳]或MRI,胸部之CT及全身放射核種骨骼掃描)。在用CT或MRI鑑別出反應之後至少4週,根據RECIST 1.1確認軟組織反應,根據獨立中心評審在重複骨骼掃描至少6週後根據前列腺癌工作組3標準無確證之骨骼進展之跡象。 Assess tumor response: Radiographic assessment (CT [preferably] or MRI of the abdomen and pelvis, CT of the chest and whole-body radionuclide bone scan) every 8 weeks for the first 24 weeks and then every 12 weeks thereafter. Confirmation of soft tissue response according to RECIST 1.1 at least 4 weeks after identification of response with CT or MRI, with no confirmed evidence of skeletal progression according to Prostate Cancer Working Group 3 criteria after repeat bone scan at least 6 weeks after independent central review.

腫瘤基因體學 使用FoundationOne ®CDx (Foundation Medicine, Inc.)在福爾馬林固定的石蠟包埋(FFPE)腫瘤組織中評估DDR11突變。gLOH分數判定為FoundationOne ®CDx分析之晚期基因體分析(方法詳述於Sokol等人, JCO Precision Oncology 2020;4:442-465中)。 Tumor Genomics : DDR11 mutations were assessed in formalin-fixed paraffin-embedded (FFPE) tumor tissue using FoundationOne ® CDx (Foundation Medicine, Inc.). The gLOH score was determined by late genome analysis of the FoundationOne ® CDx assay (methods detailed in Sokol et al., JCO Precision Oncology 2020;4:442-465).

統計分析 可評估抗腫瘤活性之病患群體(N=104)定義為根據研究人員評定在篩選時具有可量測軟組織疾病、在11個預定義DDR-HRR基因(ATM、ATR、BRCA1、BRCA2、CHEK2、FANCA、MLH1、MRE11A、NBN、PALB2及RAD51C)內具有基因改變且已接受至少一次劑量之他拉唑帕尼的所有已登記病患。對在基線處具有可量測疾病且在基線之後具有至少一個有效評定之病患依據盲態獨立中心評審評定以靶病灶之直徑總和計之相對於基線的最佳變化(作為客觀反應率結果之一部分)。依據RECIST版本1.1,腫瘤反應分類為完全反應(CR)、部分反應(PR)、疾病穩定(SD)或疾病進展(PD)。藉由盲態獨立中心評審,確認主要終點為客觀反應率(ORR),根據RECIST 1.1定義為完全反應或部分反應之最佳總體軟組織反應。次要終點包括達到客觀反應的時間、客觀反應的持續時間、相對於基線PSA減少50%或更多的病患之比例、達到PSA進展的時間、具有循環腫瘤細胞計數之轉換的病患之比例(相對於基線在任何時間每7.5 mL血液減少≥5至<5個細胞或每7.5 mL血液減少≥1至0個細胞,或自每7.5 mL血液<5個細胞之任何增加),放射無進展存活期中之進展(從第一劑量之他拉唑帕尼至軟組織(如根據RECIST 1.1放射照相術、根據盲態獨立中心評審及研究人員評定判定)、骨骼中之進展(根據前列腺癌工作組3標準及獨立中心評審)、或因任何原因之死亡,以兩者首先發生者為準)、總存活期、安全性、病患報告的結果及藥代動力學。 Statistical analysis : The patient population evaluable for anti-tumor activity (N=104) was defined as those with measurable soft tissue disease at screening, as assessed by investigators, and 11 predefined DDR-HRR genes (ATM, ATR, BRCA1, BRCA2 All enrolled patients with genetic alterations in , CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2 and RAD51C) who have received at least one dose of talazopanib. Patients with measurable disease at baseline and at least one valid assessment after baseline were assessed by blinded independent central review as the best change from baseline in the sum of diameters of target lesions (as an objective response rate outcome). part). According to RECIST version 1.1, tumor response was classified as complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD). Through blinded independent central review, the primary endpoint was confirmed to be objective response rate (ORR), defined as the best overall soft tissue response of complete response or partial response according to RECIST 1.1. Secondary endpoints include time to objective response, duration of objective response, proportion of patients with a 50% or greater reduction in PSA from baseline, time to PSA progression, and proportion of patients with conversion of circulating tumor cell count (A decrease of ≥5 to <5 cells per 7.5 mL of blood or a decrease of ≥1 to 0 cells per 7.5 mL of blood at any time from baseline, or any increase from <5 cells per 7.5 mL of blood), no progression from radiation Progression in survival (from first dose of talazopanib to soft tissue (as determined by RECIST 1.1 radiography, blinded independent central review and investigator assessment), progression in bone (as determined by Prostate Cancer Working Group 3 standard and independent center review), or death from any cause, whichever occurs first), overall survival, safety, patient-reported outcomes and pharmacokinetics.

藉由HRR基因改變組(BRCA1、BRCA2、PALB2、ATM及11個DDR-HRR基因之預定義組中的其他基因)分析事後抗腫瘤活性終點,其中使用階層式策略,藉由HRR基因改變將病患分開,其中BRCA1或BRCA2排名高於PALB2、PALB2排名高於ATM,且ATM排名高於所有其他改變。Post hoc anti-tumor activity endpoints were analyzed by the HRR gene alteration panel (BRCA1, BRCA2, PALB2, ATM and other genes in a predefined set of 11 DDR-HRR genes), using a hierarchical strategy in which the disease was differentiated by HRR gene alterations. Patients were separated in which BRCA1 or BRCA2 ranked higher than PALB2, PALB2 ranked higher than ATM, and ATM ranked higher than all other changes.

結果 抗腫瘤功效群體之病患特徵 (N=104) 9名(9%)病患為格里森組1,31名(30%)病患為格里森組2,63名(61%)病患為格里森組3,且1名(1%)病患未報告格里森組別[格里森等級及相關等級分組(1至5)係指當相比於正常前列腺細胞時癌細胞看起來如何。格里森等級組愈高,癌症將愈可能生長及擴散]。36名(35%)病患患有內臟疾病且68名(65%)病患患有非內臟疾病。初次診斷時病患之初始M階段為M0 39名(38%)、M1 48名(46%)、MX 13名(13%)且4名(4%)病患未報告[M類別捕捉癌症是否已轉移至身體之其他部分。M0:癌症尚未擴散至身體之其他部分。M1:癌症已擴散至身體之其他部分。MX:不能量測到轉移]。病患之先前紫杉烷使用如下:54名(52%)病患僅使用多西他賽,49名(47%)病患使用多西他賽及卡巴他賽,且1名(1%)病患未報告先前紫杉烷使用。病患之先前激素療法之使用如下:37名(36%)病患僅使用阿比特龍,37名(36%)病患僅使用恩雜魯胺,28名(27%)病患使用阿比特龍及恩雜魯胺,且2名(2%)病患之先前激素療法未報告。 Results Patient characteristics of the anti-tumor efficacy group (N=104) : 9 (9%) patients were in Gleason group 1, 31 (30%) patients were in Gleason group 2, and 63 (61%) patients were in Gleason group 2 Patients were Gleason group 3, and 1 (1%) patient did not report Gleason group [Gleason grade and related grade groups (1 to 5) refer to how cancer cells look when compared to normal prostate cells how. The higher the Gleason grade group, the more likely the cancer will grow and spread]. Thirty-six (35%) patients had visceral disease and 68 (65%) patients had non-visceral disease. The patients' initial M stage at initial diagnosis was M0 in 39 (38%), M1 in 48 (46%), MX in 13 (13%), and 4 (4%) patients did not report [M category captures whether the cancer Has been transferred to other parts of the body. M0: The cancer has not spread to other parts of the body. M1: The cancer has spread to other parts of the body. MX: No transfer detected]. Patients' prior taxane use was as follows: 54 (52%) patients were on docetaxel only, 49 (47%) were on both docetaxel and cabazitaxel, and 1 (1%) was on docetaxel The patient reported no prior taxane use. Patients' prior hormone therapy use was as follows: 37 (36%) patients used abiraterone only, 37 (36%) patients only used enzalutamide, and 28 (27%) patients used abiraterone only. and enzalutamide, and prior hormonal therapy was not reported in 2 (2%) patients.

55名病患(53%)可評估gLOH,45名不可評估gLOH,且四名缺乏中央實驗室gLOH結果(gLOH之可評估性反映組織純度及/或品質不足)。Fifty-five patients (53%) were evaluable for gLOH, 45 were not evaluable for gLOH, and four lacked central laboratory gLOH results (gLOH evaluability reflects insufficient tissue purity and/or quality).

他拉唑帕尼功效: 在16.4個月之中值追蹤之後,客觀反應率係29.8% (104名病患之31名;95%置信區間(CI) 21.2-39.6)。 Talazopanib Efficacy : After a median follow-up of 16.4 months, the objective response rate was 29.8% (31 of 104 patients; 95% confidence interval (CI) 21.2-39.6).

腫瘤 gLOH 分數作為他拉唑帕尼反應之預測性生物標記的評估 gLOH高/低狀態與他拉唑帕尼反應之潛在關聯性經探究且結果呈現於下表1及表2中。已探究兩個gLOH高/低臨限值。8.8%係基於證明此臨限值區分攜帶BRCA雙對偶基因突變之前列腺癌與具有最佳敏感性及特異性之BRCA野生型前列腺癌的公開案(Sokol等人,JCO Precision Oncology 2020;4:442-465)(結果呈現於表1中)選擇。此外,探索基於功效群體中之中值gLOH分數(9.2%)的不可知臨限值(結果呈現於表2中)。基於8.8%臨限值,與gLOH-低[3/25(12.0%)、95% CI 2.5,31.2%;幾率比8.381,雙側p值0.0017,基於費希爾精確檢驗(Fisher's exact test)]相比,gLOH-高[16/30(53.3%),95% CI,根據克洛珀-皮爾森方法(Clopper-Pearson method) 34.3,71.7%)]之ORR顯著較高。基於9.2% gLOH臨限值,相對於gLOH-低[4/27(14.8%);95% CI 4.2,33.7;幾率比6.635,p值0.0041],gLOH-高[15/28(53.6%),95% CI 33.9,72.5%]之ORR亦顯著較高。選擇8.8%臨限值用於下文詳述之其他gLOH高/低分析。 Evaluation of tumor gLOH score as a predictive biomarker of talazopanib response : The potential association of high/low gLOH status with talazopanib response was explored and the results are presented in Tables 1 and 2 below. Two gLOH high/low thresholds have been explored. 8.8% is based on published studies demonstrating that this cutoff differentiates prostate cancer harboring BRCA doublet mutations from BRCA wild-type prostate cancer with optimal sensitivity and specificity (Sokol et al., JCO Precision Oncology 2020;4:442 -465) (results presented in Table 1) selection. Additionally, an agnostic threshold based on the median gLOH score in the efficacy population (9.2%) was explored (results are presented in Table 2). Based on 8.8% cutoff, compared with gLOH-low [3/25 (12.0%), 95% CI 2.5, 31.2%; odds ratio 8.381, two-sided p-value 0.0017, based on Fisher's exact test] In comparison, the ORR was significantly higher for gLOH-high [16/30 (53.3%), 95% CI, 34.3, 71.7% by Clopper-Pearson method). Based on 9.2% gLOH cutoff, relative to gLOH-low [4/27 (14.8%); 95% CI 4.2, 33.7; odds ratio 6.635, p value 0.0041], gLOH-high [15/28 (53.6%), The ORR (95% CI 33.9, 72.5%] was also significantly higher. The 8.8% threshold was selected for use in other gLOH high/low analyzes detailed below.

概言之,使用任一臨限值,相對於gLOH-低,gLOH-高之ORR顯著較高,儘管gLOH-低病患中針對兩個臨限值均存在反應。 1 基於 8.8% 臨限值藉由 gLOH / 低狀態基於 BICR 評估 (RECIST 1.1 PCWG3) 之反應 (gLOH 之可評估功效群體 N=55) 反應 gLOH- (n=30) gLOH- (n=25) 確認最佳整體反應,n(%) 完全反應(CR) 3 (10.0) 1 (4.0) 部分反應(PR) 13 (43.3) 2 (8.0) 疾病穩定(SD) 6 (20.0) 11 (44.0) 非CR/非PD 2 (6.7) 0 疾病進展(PD) 4 (13.3) 7 (28.0) 不可評估(NE) 2 (6.7) 4 (16.0) 客觀反應 (CR+PR) n (% [95% CI a]) 16 (53.3 [34.3,71.7]) 3 (12.0 [2.5,31.2]) ORR比較對比gLOH-低之非分層分析 幾率比 (95% CI b) 8.381 (1.833,50.950)    雙側P值 c 0.0017    a使用的克洛珀-皮爾森方法; b幾率比>1指示相比於低,高之結果較好;計算出準確CI。 c基於費希爾精確檢驗之P值。 2 基於 9.2% 臨限值藉由 gLOH / 低狀態基於 BICR 評估 (RECIST 1.1 PCWG3) 之反應 (gLOH 之可評估功效群體 N=55) 反應 gLOH- (n=28) gLOH- (n=27) 確認最佳整體反應,n(%) 完全反應(CR) 2 (7.1) 2 (7.4) 部分反應(PR) 13 (46.4) 2 (7.4) 疾病穩定(SD) 5 (17.9) 12 (44.4) 非CR/非PD 2 (7.1) 0 疾病進展(PD) 4 (14.3) 7 (25.9) 不可評估(NE) 2 (7.1) 4 (14.8) 客觀反應 (CR+PR), n (% [95% CI a]) 15 (53.6 [33.9,72.5]) 4 (14.8 [4.2,33.7]) ORR比較對比gLOH-低之非分層分析 幾率比(95% CI b) 6.635 (1.596,32.224)    雙側P值 c 0.0041    a使用的克洛珀-皮爾森方法; b幾率比>1指示相比於低,高之結果較好;計算出準確CI。 c基於費希爾精確檢驗之P值。 In summary, using either cutoff, gLOH-high had a significantly higher ORR relative to gLOH-low, although responses were seen in gLOH-low patients at both cutoffs. Table 1 : Response ( evaluable efficacy population for gLOH , N=55) based on BICR assessment (RECIST 1.1 ; PCWG3) by gLOH high / low status based on 8.8% threshold . reaction gLOH- High (n=30) gLOH- low (n=25) Confirm best overall response, n(%) Complete response (CR) 3 (10.0) 1 (4.0) Partial response (PR) 13 (43.3) 2 (8.0) Stable disease (SD) 6 (20.0) 11 (44.0) Non-CR/Non-PD 2 (6.7) 0 Progressive disease (PD) 4 (13.3) 7 (28.0) Not evaluable (NE) 2 (6.7) 4 (16.0) Objective response (CR+PR) , n (% [95% CI a ]) 16 (53.3 [34.3,71.7]) 3 (12.0 [2.5,31.2]) ORR comparison versus gLOH-low non-stratified analysis Odds ratio (95% CI b ) 8.381 (1.833,50.950) Two-sided P value c 0.0017 a The Clopper-Pearson method used; b Odds ratio >1 indicates that higher results are better than lower ones; accurate CI is calculated. c P value based on Fisher’s exact test. Table 2 : Response based on BICR assessment (RECIST 1.1 ; PCWG3) by gLOH high / low status based on 9.2% cutoff (gLOH evaluable efficacy population , N=55) reaction gLOH- High (n=28) gLOH- low (n=27) Confirm best overall response, n(%) Complete response (CR) 2 (7.1) 2 (7.4) Partial response (PR) 13 (46.4) 2 (7.4) Stable disease (SD) 5 (17.9) 12 (44.4) Non-CR/Non-PD 2 (7.1) 0 Progressive disease (PD) 4 (14.3) 7 (25.9) Not evaluable (NE) 2 (7.1) 4 (14.8) Objective response (CR+PR), n (% [95% CI a ]) 15 (53.6 [33.9,72.5]) 4 (14.8 [4.2,33.7]) ORR comparison versus gLOH-low non-stratified analysis Odds ratio (95% CI b ) 6.635 (1.596,32.224) Two-sided P valuec 0.0041 a The Clopper-Pearson method used; b Odds ratio >1 indicates that higher results are better than lower ones; accurate CI is calculated. c P value based on Fisher’s exact test.

隨後,使用藉由最佳反應、突變組、突變類型及突變接合子型式標記之gLOH之條形圖探究gLOH分數與基因突變組內之反應的潛在關聯(圖式)。Subsequently, potential correlations between gLOH scores and responses within gene mutation groups were explored using bar plots of gLOH marked by best response, mutation group, mutation type, and mutation adapter pattern (Graph).

來自此分析之所選結果列於表3中。 3 藉由基因突變組及 gLOH / 低狀態根據 RECIST v.1.1 之最佳總體反應 (gLOH 可評估之功效群體 N=55) 基因 #gLOH- #gLOH- ORR (%)(gLOH- 低) ORR (%)(gLOH- 高) BRCA1 (n=2) 0 2 (100%) - 2/2 (100%) BRCA2 (n=30) 13 (43%) 17 (57%) 3/13 (23%) 12/17 (71%) PALB2 (n=2) 2 (100%) 0 0/2 (0%) - ATM (n=10) 6 (60%) 4 (40%) 0/6 (0%) 2/4 (50%) 其他(n=11) 4 (36%) 7 (64%) 0/4 (0%) 0/7 (0%) Selected results from this analysis are listed in Table 3. Table 3 : Best overall response according to RECIST v.1.1 by gene mutation group and gLOH high / low status (gLOH evaluable efficacy population , N=55) Gene #gLOH- low #gLOH- High ORR (%)(gLOH- Low) ORR (%)(gLOH- High) BRCA1 (n=2) 0 2 (100%) - 2/2 (100%) BRCA2 (n=30) 13 (43%) 17 (57%) 3/13 (23%) 12/17 (71%) PALB2 (n=2) 2 (100%) 0 0/2 (0%) - ATM (n=10) 6 (60%) 4 (40%) 0/6 (0%) 2/4 (50%) Others(n=11) 4 (36%) 7 (64%) 0/4 (0%) 0/7 (0%)

如表4中所示,在BRCA2突變組內,不管gLOH狀態如何,ORR為穩定的,但對於gLOH-高(12/17,70.6%),gLOH-高之ORR顯著高於gLOH-低(3/13,23.1%) [根據費希爾精確檢驗p=0.0253]。 4 基於 8.8% 臨限值 gLOH 狀態與對攜帶 BRCA2 改變之腫瘤之反應的關聯 反應 BRCA2 gLOH- (n=17) gLOH- (n=13) 確認最佳整體反應,n(%) 完全反應(CR) 2 (11.8) 1 (7.7) 部分反應(PR) 10 (58.8) 2 (15.4) 疾病穩定(SD) 2 (11.8) 7 (53.8) 非CR/非PD 2 (11.8) 0 疾病進展(PD) 0 1 (7.7) 不可評估(NE) 1 (5.9) 2 (15.4) 客觀反應 (CR+PR) n (% [95% CI a]) 12 (70.6 [44.0,89.7]) 3 (23.1 [5.0,53.8]) ORR比較對比gLOH-低之非分層分析 幾率比(95% CI b) 8.000 (1.220,60.952)    雙側P值 c 0.0253    a使用的克洛珀-皮爾森方法; b幾率比>1指示相比於低,高之結果較好;計算出準確CI; c基於費希爾精確檢驗之P值 As shown in Table 4, within the BRCA2 mutation group, the ORR was stable regardless of gLOH status, but for gLOH-high (12/17, 70.6%), the ORR of gLOH-high was significantly higher than that of gLOH-low (3 /13, 23.1%) [p=0.0253 according to Fisher's exact test]. Table 4 : Association of gLOH status and response to tumors harboring BRCA2 alterations based on 8.8% cutoff reaction BRCA2 gLOH- High (n=17) gLOH- low (n=13) Confirm best overall response, n(%) Complete response (CR) 2 (11.8) 1 (7.7) Partial response (PR) 10 (58.8) 2 (15.4) Stable disease (SD) 2 (11.8) 7 (53.8) Non-CR/Non-PD 2 (11.8) 0 Progressive disease (PD) 0 1 (7.7) Not evaluable (NE) 1 (5.9) 2 (15.4) Objective response (CR+PR) , n (% [95% CI a ]) 12 (70.6 [44.0,89.7]) 3 (23.1 [5.0,53.8]) ORR comparison versus gLOH-low non-stratified analysis Odds ratio (95% CI b ) 8.000 (1.220,60.952) Two-sided P value c 0.0253 a The Clopper-Pearson method used; b Odds ratio >1 indicates that high results are better than low ones; accurate CI is calculated; c P value based on Fisher's exact test

如表5中所示,在ATM突變子組內,對於gLOH-高(2/4,50%),gLOH-高之ORR在數值上高於gLOH-低(0/6,0%),但並不顯著(P=0.1333)。 5 基於 8.8% 臨限值之 gLOH 狀態與對攜帶 ATM 改變之腫瘤之反應的關聯 反應 ATM gLOH- (n=4) gLOH- (n=6) 確認最佳整體反應,n(%) 完全反應(CR) 1 (25.0) 0 部分反應(PR) 1 (25.0) 0 疾病穩定(SD) 1 (25.0) 3 (50.0) 非CR/非PD 0 0 疾病進展(PD) 1 (25.0) 3 (50.0) 不可評估(NE) 0 0 客觀反應 (CR+PR) n (% [95% CI a]) 2 (50.0 [6.8,93.2]) 0 (0 [0,45.9]) ORR比較對比gLOH-低之非分層分析 幾率比(95% CI b) NE    雙側P值 c 0.1333    a使用的克洛珀-皮爾森方法; b幾率比>1指示相比於低,高之結果較好;計算出準確CI; c基於費希爾精確檢驗之P值 As shown in Table 5, within the ATM mutant subgroup, the ORR for gLOH-high (2/4, 50%) was numerically higher than for gLOH-low (0/6, 0%), but Not significant (P=0.1333). Table 5 : Association of gLOH status and response to tumors harboring ATM alterations based on 8.8% cutoff reaction ATM gLOH- High (n=4) gLOH- low (n=6) Confirm best overall response, n(%) Complete response (CR) 1 (25.0) 0 Partial response (PR) 1 (25.0) 0 Stable disease (SD) 1 (25.0) 3 (50.0) Non-CR/Non-PD 0 0 Progressive disease (PD) 1 (25.0) 3 (50.0) Not evaluable (NE) 0 0 Objective response (CR+PR) , n (% [95% CI a ]) 2 (50.0 [6.8,93.2]) 0 (0 [0,45.9]) ORR comparison versus gLOH-low non-stratified analysis Odds ratio (95% CI b ) NE Two-sided P value c 0.1333 a The Clopper-Pearson method used; b Odds ratio >1 indicates that high results are better than low ones; accurate CI is calculated; c P value based on Fisher's exact test

使用任一臨限值(風險比0.68),gLOH可評估功效群體中之放射無進展存活期(RECIST 1.1;BICR)在數值上gLOH-高優於gLOH-低,但並不顯著。基於8.8%臨限值之結果列於表6中。表6中所示之gLOH-高與gLOH-低之早期分離的標誌隨後缺失。9.2%臨限值之風險比(95% CI b)為0.68 (0.317,1.448);雙側P值=0.3131(詳細結果未示出)。 6 基於 8.8% 臨限值藉由 gLOH / 低狀態之放射無進展存活期 (RECIST 1.1 BICR)( 可評估 gLOH 之功效群體 N=55) gLOH- (n=30) gLOH- (n=25) 至事件之時間的卡本-麥爾估算(Kaplan-Meier estimates),月(95% CI) a 四分位數1 5.4 (1.7,8.3) 1.8 (1.6,3.7) 中值 10.9 (7.5,NE) 11.1 (2.1,19.2) 四分位數3 NE (19.3,NE) 19.2 (11.1,19.2) rPFS比較對比gLOH-低之非分層分析 風險比(95% CI b) 0.68 (0.319,1.456)    雙側P值 c 0.3219    a基於布絡克菲亞及克勞來方法(Brookmeyer and Crowley method); b基於Cox比例風險模型的風險比;在比例風險下,風險比<1指示相較於gLOH-低,風險率之降低有利於gLOH-高; c基於對數等級檢定之P值; Radiation progression-free survival (RECIST 1.1; BICR) in the gLOH-evaluable efficacy population was numerically, but not significantly, better for gLOH-high than gLOH-low using either cutoff (hazard ratio 0.68). The results based on the 8.8% threshold are listed in Table 6. Signs of the early separation of gLOH-high and gLOH-low shown in Table 6 were subsequently missing. The risk ratio (95% CI b ) of the 9.2% threshold was 0.68 (0.317, 1.448); two-sided P value = 0.3131 (detailed results are not shown). Table 6 : Radiation progression-free survival by gLOH high / low status based on 8.8% cutoff (RECIST 1.1 ; BICR) ( gLOH efficacy evaluable population , N=55) . gLOH- High (n=30) gLOH- low (n=25) Kaplan-Meier estimates of time to event, months (95% CI) a Quartile 1 5.4 (1.7,8.3) 1.8 (1.6,3.7) median 10.9 (7.5,NE) 11.1 (2.1,19.2) Quartile 3 NE (19.3,NE) 19.2 (11.1,19.2) rPFS comparison versus gLOH-low non-stratified analysis Risk ratio (95% CI b ) 0.68 (0.319,1.456) Two-sided P value c 0.3219 aBased on the Brookmeyer and Crowley method; bHazard ratio based on the Cox proportional hazard model; under proportional hazards, a hazard ratio <1 indicates a reduction in the hazard rate compared to gLOH-low. Favoring gLOH-high; c P value based on log-rank test;

因此,gLOH-高狀態與功效群體內之反應相關聯且亦與BRCA2基因突變組內之反應相關聯。Thus, gLOH-high status was associated with response within the efficacy group and also with response within the BRCA2 gene mutation group.

隨後,使用相同的經標註條形圖探究gLOH分數與基因接合子型式及突變類型子組內反應之潛在關聯(圖式)。基於此觀測,顯然接合子型式與gLOH之間無明顯關係。關於突變類型,DDR11短變體(亦即單核苷酸變體、短嵌入/缺失)廣泛地分佈在gLOH範圍內。相比之下,複本數缺失(其受限於BRCA2基因改變子組;n=13 pts)在數值上與高gLOH分數及反應相關聯:3/25 gLOH-低具有BRCA2複本數缺失(2 SD,1 NE),而10/30 gLOH-高具有BRCA2複本數缺失(8 CR/PR,1 SD,1非CR/PD)。此表明DDR11m-gLOH-高相對於DDR11m-gLOH-低之統計上優良總體功效(53%相對於12%,p=0.0017)且BRCA2m-gLOH-高相對於BRCA2m-gLOH-低之優良功效(71%相對於23%,p=0.0253)在某種程度上反映在gLOH-高組中之BRCA2複本數缺失之相對高分數。The same annotated bar graphs were then used to explore potential correlations between gLOH scores and responses within subgroups of gene zygote patterns and mutation types (Figure). Based on this observation, it is apparent that there is no clear relationship between zygote pattern and gLOH. Regarding mutation types, DDR11 short variants (i.e., single nucleotide variants, short insertions/deletions) are widely distributed across the gLOH spectrum. In contrast, copy number deletions (which were restricted to a subset of BRCA2 gene alterants; n=13 pts) were numerically associated with high gLOH scores and responses: 3/25 gLOH-low had BRCA2 copy number deletions (2 SD , 1 NE), whereas 10/30 gLOH-high had BRCA2 copy number deletions (8 CR/PR, 1 SD, 1 non-CR/PD). This demonstrates statistically superior overall efficacy of DDR11m-gLOH-high versus DDR11m-gLOH-low (53% versus 12%, p=0.0017) and superior efficacy of BRCA2m-gLOH-high versus BRCA2m-gLOH-low (71 % vs. 23%, p=0.0253) was reflected to some extent by the relatively high fraction of BRCA2 copy number deletions in the gLOH-high group.

基於此大量預治療之mCRPC群體中之此等回溯性特用探索性分析,gLOH-高狀態與在gLOH可評估功效群體內及在gLOH可評估 BRCA2改變子組內對他拉唑帕尼之增強的反應相關聯。此等結果證明在DDR11m前列腺癌中及亦在11個預定義DDR-HRR基因改變子組中之一些內針對gLOH-高狀態之預測潛力。 Based on these retrospective ad hoc exploratory analyzes in this heavily pretreated mCRPC population, gLOH-high status was associated with enhancement of talazopanib within the gLOH-evaluable efficacy population and within the gLOH-evaluable BRCA2 alteration subgroup. reaction is related. These results demonstrate the predictive potential for gLOH-high status in DDR11m prostate cancer and also within some of the 11 predefined DDR-HRR gene alteration subgroups.

總之,TALAPRO-1中之此等回溯性探索性分析證明,gLOH高狀態與在具有DNA修復基因突變之轉移性去勢抗性前列腺癌中之他拉唑帕尼的反應相關聯。In summary, these retrospective exploratory analyzes in TALAPRO-1 demonstrate that high gLOH status is associated with response to talazopanib in metastatic castration-resistant prostate cancer with DNA repair gene mutations.

本說明書中所引用之所有公開案及專利申請案均以全文引用之方式併入本文中。儘管已藉助於說明及實例相當詳細地描述前述發明,但對於一般熟習此項技術者根據本發明之教示內容顯而易知,在不背離隨附申請專利範圍之精神或範疇的情況下,可對其進行某些改變及修改。All publications and patent applications cited in this specification are incorporated herein by reference in their entirety. Although the foregoing invention has been described in considerable detail by way of illustration and example, it will be apparent to those skilled in the art from the teachings of the invention that the invention may be practiced without departing from the spirit or scope of the appended claims. Make certain changes and modifications thereto.

圖式顯示基於盲態獨立中心評審之gLOH分數之最佳總體反應(已確認)的條形圖。條形圖藉由最佳反應、突變組、突變類型及突變接合子型式標註。縮寫包括:CR,完全反應;PR,部分反應;SD,疾病穩定;非CR/非PD,非完全反應/非疾病進展;PD,疾病進展;NE,不可評估;gLOH (基因雜合性缺失)及alt (改變或突變)。其他DDR11基因包括但不限於ATR、CHEK2、FANCA、MLH1、MRE11A、NBN及RAD51C。腫瘤變體類型係基於相應DDR11突變組。關於接合子型式之定義包括:同型接合,病患具有≥1同型接合DDR11突變;異型接合,病患不具有同型接合DDR11突變,但具有≥1異型接合DDR11突變;未知,病患不具有同型接合或異型接合DDR11突變,但具有≥1未知接合子型式之DDR11突變;不可評估,病患不具有充分的DDR11突變。Graph showing bar chart of best overall response (confirmed) based on blinded independent center review of gLOH scores. Bar graphs are annotated by best response, mutation group, mutation type, and mutation zygote pattern. Abbreviations include: CR, complete response; PR, partial response; SD, stable disease; non-CR/non-PD, non-complete response/non-progressive disease; PD, progressive disease; NE, not evaluable; gLOH (gene loss of heterozygosity) and alt (change or mutation). Other DDR11 genes include, but are not limited to, ATR, CHEK2, FANCA, MLH1, MRE11A, NBN and RAD51C. Tumor variant types are based on corresponding DDR11 mutation sets. The definitions of the zygote pattern include: homozygous, the patient has ≥1 homozygous DDR11 mutation; heterozygous, the patient does not have homozygous DDR11 mutation, but has ≥1 heterozygous DDR11 mutation; unknown, the patient does not have homozygous DDR11 mutation or heterozygous DDR11 mutation, but with ≥1 DDR11 mutation of unknown zygote type; not evaluable, the patient does not have sufficient DDR11 mutations.

Claims (16)

一種選擇患有具有同源重組修復缺陷之癌症之個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療之方法,其包含:a)根據該癌症之活檢判定基因雜合性缺失(gLOH)分數;及b)基於該gLOH分數選擇該個體以用他拉唑帕尼或其醫藥學上可接受之鹽治療。A method of selecting individuals with cancer having homologous recombination repair deficiency for treatment with talazopanib or a pharmaceutically acceptable salt thereof, comprising: a) determining loss of gene heterozygosity based on a biopsy of the cancer (gLOH) score; and b) selecting the individual for treatment with talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score. 一種治療個體內具有同源重組修復缺陷之癌症之方法,其包含:a)根據如請求項1之方法選擇該個體;及b)基於該gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。A method of treating cancer with homologous recombination repair deficiency in an individual, comprising: a) selecting the individual according to the method of claim 1; and b) administering a therapeutically effective amount of talazopanib or based on the gLOH score Its pharmaceutically acceptable salt. 一種鑑別對以他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,其包含:a)根據該癌症之活檢判定基因雜合性缺失(gLOH)分數;及b)基於該gLOH分數選擇該癌症以用他拉唑帕尼或其醫藥學上可接受之鹽治療。A method of identifying cancers with homologous recombination repair defects that are sensitive to treatment with talazopanib or a pharmaceutically acceptable salt thereof, comprising: a) determining gene loss of heterozygosity (gLOH) based on a biopsy of the cancer ) score; and b) selecting the cancer for treatment with talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score. 一種治療對以他拉唑帕尼或其醫藥學上可接受之鹽治療敏感的具有同源重組修復缺陷之癌症之方法,其包含:a)根據如請求項3之方法選擇該癌症;及b)基於該gLOH分數投與治療有效量之他拉唑帕尼或其醫藥學上可接受之鹽。A method of treating cancer with homologous recombination repair deficiency that is sensitive to treatment with talazopanib or a pharmaceutically acceptable salt thereof, comprising: a) selecting the cancer according to the method of claim 3; and b ) administer a therapeutically effective amount of talazopanib or a pharmaceutically acceptable salt thereof based on the gLOH score. 如請求項1或請求項2之方法,其中該癌症對以他拉唑帕尼治療敏感。The method of claim 1 or claim 2, wherein the cancer is sensitive to treatment with talazopanib. 如請求項1至4中任一項之方法,其中該癌症係前列腺癌。The method of any one of claims 1 to 4, wherein the cancer is prostate cancer. 如請求項6之方法,其中該前列腺癌係轉移性前列腺癌。The method of claim 6, wherein the prostate cancer is metastatic prostate cancer. 如請求項6之方法,其中該前列腺癌係去勢抗性前列腺癌。The method of claim 6, wherein the prostate cancer is castration-resistant prostate cancer. 如請求項8之方法,其中該去勢抗性前列腺癌係轉移性去勢抗性前列腺癌。The method of claim 8, wherein the castration-resistant prostate cancer is metastatic castration-resistant prostate cancer. 如請求項1或請求項3之方法,其中該癌症之同源重組修復缺陷藉由下一代定序判定。The method of claim 1 or claim 3, wherein the homologous recombination repair defect of the cancer is determined by next-generation sequencing. 如請求項1或請求項3之方法,其中步驟a)係藉由下一代定序進行。The method of claim 1 or claim 3, wherein step a) is performed by next-generation sequencing. 如請求項1至11中任一項之方法,其中該gLOH分數係至少約8.0%。The method of any one of claims 1 to 11, wherein the gLOH fraction is at least about 8.0%. 如請求項1至11中任一項之方法,其中該gLOH分數係至少約8.3%。The method of any one of claims 1 to 11, wherein the gLOH fraction is at least about 8.3%. 如請求項1至11中任一項之方法,其中該gLOH分數係至少約8.8%。The method of any one of claims 1 to 11, wherein the gLOH fraction is at least about 8.8%. 如請求項1至11中任一項之方法,其中該gLOH分數係至少約9%;至少約9.2%;至少約10%;至少約11%;至少約12%;至少約13%;至少約14%;至少約15%;至少約16%;至少約17%;至少約18%;至少約19%;至少約20%;至少約21%;至少約22%;至少約23%;至少約24%;或至少約25%。The method of any one of claims 1 to 11, wherein the gLOH fraction is at least about 9%; at least about 9.2%; at least about 10%; at least about 11%; at least about 12%; at least about 13%; at least about 14%; at least about 15%; at least about 16%; at least about 17%; at least about 18%; at least about 19%; at least about 20%; at least about 21%; at least about 22%; at least about 23%; at least about 24%; or at least about 25%. 如請求項1或2之方法,其中該個體係人類。 Such as the method of claim 1 or 2, wherein the system is human.
TW112100665A 2022-01-08 2023-01-07 Genomic loss of heterozygosity as a predictive biomarker for treatment with talazoparib and methods of treatment thereof TW202339754A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263297741P 2022-01-08 2022-01-08
US63/297,741 2022-01-08
US202263321170P 2022-03-18 2022-03-18
US63/321,170 2022-03-18

Publications (1)

Publication Number Publication Date
TW202339754A true TW202339754A (en) 2023-10-16

Family

ID=84943502

Family Applications (1)

Application Number Title Priority Date Filing Date
TW112100665A TW202339754A (en) 2022-01-08 2023-01-07 Genomic loss of heterozygosity as a predictive biomarker for treatment with talazoparib and methods of treatment thereof

Country Status (2)

Country Link
TW (1) TW202339754A (en)
WO (1) WO2023131894A1 (en)

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG193842A1 (en) 2008-08-06 2013-10-30 Biomarin Pharm Inc Dihydropyridophthalazinone inhibitors of poly(adp-ribose)polymerase (parp)
JP5883397B2 (en) 2010-02-03 2016-03-15 ビオマリン プハルマセウトイカル インコーポレイテッド Use of dihydropyridphthalazinone inhibitors of poly (ADP-ribose) polymerase (PARP) in the treatment of diseases associated with PTEN deficiency
KR101826652B1 (en) 2010-02-08 2018-02-07 메디베이션 테크놀로지즈, 인크. Processes of synthesizing dihydropyridophthalazinone derivatives
PT2630146T (en) 2010-10-21 2020-07-20 Medivation Tech Llc Crystaline (8s,9r)-5-fluoro-8-(4-fluorophenyl)-9-(1-methyl-1h-1,2,4-triazol-5-yl)-8,9-dihydro-2h-pyrido[4,3,2-de]phtalazin-3(7h)-one tosylate salt
WO2015069851A1 (en) 2013-11-07 2015-05-14 Biomarin Pharmaceutical Inc. Triazole intermediates useful in the synthesis of protected n-alkyltriazolecarbaldehydes
ES2947501T3 (en) 2014-07-31 2023-08-10 Medivation Tech Llc Coforming salts of (2S,3S)-methyl 7-fluoro-2-(4-fluorophenyl)-3-(1-methyl-1h-1,2,4-triazol-5-yl)-4-oxo-1, 2,3,4-tetrahydroquinoline-5-carboxylate and processes for preparing it
WO2017075091A1 (en) 2015-10-26 2017-05-04 Medivation Technologies, Inc. Treatment of small cell lung cancer with a parp inhibitor
AU2018394976A1 (en) * 2017-12-27 2020-07-16 Tesaro, Inc. Methods of treating cancer

Also Published As

Publication number Publication date
WO2023131894A1 (en) 2023-07-13

Similar Documents

Publication Publication Date Title
JP7083760B2 (en) How to treat ovarian cancer
US11959142B2 (en) Detection of cancer
US20230210852A1 (en) Methods of treating cancer in patients with an anomalous kras gene or deletions within chromosome 9
CA2990703A1 (en) Biomarkers for nanoparticle compositions
US9255270B2 (en) Methods used in sensitizing invasive glioblastoma to therapeutic treatment
JP2022553041A (en) Methods of treating HER2-positive breast cancer with tucatinib in combination with capecitabine and trastuzumab
US20170342503A1 (en) Xrn2 as a determinant of sensitivity to dna damage
US20200397796A1 (en) Methods of treatment of cancer comprising chk1 inhibitors
US20180057888A1 (en) Kub5/hera as a determinant of sensitivity to dna damage
US20210251998A1 (en) Systems and methods for treating cancer
WO2019200252A1 (en) Non-invasive detection of response to immunotherapy
WO2016172517A1 (en) Methods of treating prostate cancer
TW202339754A (en) Genomic loss of heterozygosity as a predictive biomarker for treatment with talazoparib and methods of treatment thereof
US20220040173A1 (en) Methods of delaying pain progression and treating prostate cancer
US20220226338A1 (en) Methods of Treating Cancer Using CHK1 Inhibitors
US20240052423A1 (en) Methods of identifying a tumor that is sensitive to treatment with talazoparib and methods of treatment thereof
US20210079384A1 (en) Non-invasive detection of response to a targeted therapy
US11149300B1 (en) Methods of treating gastrointestinal malignancies
Shetty et al. Molecular target therapy for gastrointestinal stromal tumors
US20230241063A1 (en) Methods of treating prostate cancer
WO2024088275A1 (en) Use of naphthylamide compound in treating drug-resistant tumors
Smyth et al. Chapter 0 Principles of Oncology and Palliative Care
WO2023159066A1 (en) Use of niraparib for the treatment of brain cancer
Jane Lim-Fat et al. PATH-03. CLINICAL UTILITY OF NEXT GENERATION SEQUENCING IN IDH-WILDTYPE GLIOBLASTOMA: THE DANA-FARBER CANCER INSTITUTE EXPERIENCE
WO2022229846A1 (en) Treatment of cancer using a transforming growth factor beta receptor type 1 inhibitor