佳學(xué)基因遺傳病基因檢測(cè)機(jī)構(gòu)排名,三甲醫(yī)院的選擇

基因檢測(cè)就找佳學(xué)基因!

熱門搜索
  • 癲癇
  • 精神分裂癥
  • 魚鱗病
  • 白癜風(fēng)
  • 唇腭裂
  • 多指并指
  • 特發(fā)性震顫
  • 白化病
  • 色素失禁癥
  • 狐臭
  • 斜視
  • 視網(wǎng)膜色素變性
  • 脊髓小腦萎縮
  • 軟骨發(fā)育不全
  • 血友病

客服電話

4001601189

在線咨詢

CONSULTATION

一鍵分享

CLICK SHARING

返回頂部

BACK TO TOP

分享基因科技,實(shí)現(xiàn)人人健康!
×
查病因,阻遺傳,哪里干?佳學(xué)基因準(zhǔn)確有效服務(wù)好! 靶向用藥怎么搞,佳學(xué)基因測(cè)基因,優(yōu)化療效 風(fēng)險(xiǎn)基因哪里測(cè),佳學(xué)基因
當(dāng)前位置:????致電4001601189! > 解決方案 > 靶向藥物 >

【佳學(xué)基因檢測(cè)】基因檢測(cè)指導(dǎo)髓母細(xì)胞瘤的代謝治療

【佳學(xué)基因】基因檢測(cè)指導(dǎo)髓母細(xì)胞瘤的代謝治療 根據(jù)《個(gè)性化腫瘤治療的方法與途徑》,與表觀遺傳學(xué)、轉(zhuǎn)錄組學(xué)和基因組學(xué)相比,髓母細(xì)胞瘤代謝組學(xué)是一個(gè)仍處于起步階段的領(lǐng)域。因此

佳學(xué)基因檢測(cè)】基因檢測(cè)指導(dǎo)髓母細(xì)胞瘤的代謝治療

根據(jù)《個(gè)性化腫瘤治療的方法與途徑》,與表觀遺傳學(xué)、轉(zhuǎn)錄組學(xué)和基因組學(xué)相比,髓母細(xì)胞瘤代謝組學(xué)是一個(gè)仍處于起步階段的領(lǐng)域。因此,針對(duì)髓母細(xì)胞瘤的代謝和非WNT/non-SHH 髓母細(xì)胞瘤的特異性治療概念很少。一種方法是抑制IDO1。髓母細(xì)胞瘤的致病基因鑒定基因解碼表明,IDO1是一種主要參與色氨酸分解代謝的酶,已被確定為創(chuàng)造免疫抑制微環(huán)境的關(guān)鍵角色[91]。一項(xiàng)小型研究報(bào)告了27個(gè)髓母細(xì)胞瘤樣本隊(duì)列中IDO1的表達(dá),其中包括16名非WNT/非SHH患者[92]。去年完成了一項(xiàng)I期試驗(yàn),該試驗(yàn)將IDO1抑制劑吲哚昔莫與替莫唑胺聯(lián)合應(yīng)用于患有反復(fù)性/難治性中樞神經(jīng)系統(tǒng)惡性腫瘤的兒童群體,并納入了81名患者。雖然該試驗(yàn)的結(jié)果尚未公布,但應(yīng)用吲哚肟與放化療聯(lián)合應(yīng)用的II期研究目前已開放供參與。除色氨酸外,多胺代謝已被確定為幾種癌癥的潛在脆弱性,包括SHH髓母細(xì)胞瘤,因?yàn)槎喟穮⑴c許多細(xì)胞過程的調(diào)節(jié)。干擾多胺合成的一種可能性是使用二氟甲基鳥氨酸(DFMO/Eflornithine)抑制鳥氨酸脫羧酶(ODC),這是一種有效的ODC抑制劑,已被測(cè)試用于各種類型的癌癥,包括神經(jīng)母細(xì)胞瘤。DFMO已經(jīng)被批準(zhǔn)用于治療昏睡病和多毛癥,這使其成為一個(gè)有趣的舊藥新用的創(chuàng)新藥物候選者。然而,由于毒性問題和藥代動(dòng)力學(xué)方面有許多問題,對(duì)其使用仍存在爭議。雖然缺乏臨床前研究,但二氟甲基鳥氨酸目前正在擴(kuò)大使用環(huán)境(NCT03581240)和探索其作為高危髓母細(xì)胞瘤樣維持療法的II期試驗(yàn)中,對(duì)非WNT/非SHH 髓母細(xì)胞瘤樣患者進(jìn)行試驗(yàn)。經(jīng)過髓母細(xì)胞瘤基因解碼,與其他針對(duì)非WNT/非SHH 髓母細(xì)胞瘤樣代謝組學(xué)脆弱性的惡性腫瘤一樣,這可能是增強(qiáng)治療支柱的一種有價(jià)值的方法。迄今為止,缺乏臨床前和臨床證據(jù)不允許對(duì)這一分子亞群做出任何結(jié)論或展望。

 

2.8. Immunotherapy

 

2.8.1. Immune Checkpoint Inhibitors


Immune checkpoint inhibitors that interfere with the PD1/PDL1- and CTLA4-mediated immunosuppressive crosstalk between malignant and immune effector cells are celebrated as one of the most important developments in oncology during the last decade []. However, these strategies rely on the presence of tumor-infiltrating lymphocytes and especially the blockade of the PD1/PDL1-axis is at least partly dependent on the expression of PDL1 on the respective tumor cells. A series of studies used immunohistochemistry and bioinformatic deconvolution to assess immune infiltration and PDL1-expression in MB [,,,,,,,,]. Although most studies only analyzed small case series or cohorts of MB patients, together they suggest that MB is an immunologically “cold” tumor with only sparse immune infiltration. Furthermore, apart from one notable exception [], all studies showed negligible or no PDL1-expression at all, especially for non-WNT/non-SHH MB [,,,,,]. Additionally, studies investigating intratumoral heterogeneity using single cell RNA-sequencing reported only minor infiltration of tumor-specific lymphocytes and a diverse spectrum of myeloid cells and microglia, which potentially contribute to an immunosuppressive tumor microenvironment [,,]. These findings indicate that it may be challenging to implement immune checkpoint inhibition as part of future treatment strategies for these patients, at least in form of a monotherapy.

However, limited preclinical findings still warrant further validation of immune checkpoint blockade as a therapeutic concept for non-WNT/non-SHH MB (Figure 2), e.g., as part of a combination therapy that induces a “hotter” tumor microenvironment such as the abovementioned INFORM2 NivEnt trial [,,]. Currently, several clinical trials that offer immune checkpoint inhibitors as a monotherapy are also open to patients with recurrent or relapsed MB (NCT02359565: Pembrolizumab and NCT03173950: Nivolumab) (Table 1). However, for the latter trial only patients >18 years of age are eligible, and these patients most often do not harbor non-WNT/non-SHH MB. Another phase II trial that tested either Nivolumab as a monotherapy or in combination with Ipilimumab is in the stage of finalization (NCT03130959). Lastly, a currently running industrial trial tests the combination of Nivolumab with the immunostimulant Bempegaldesleukin, a recombinant form of IL-2, in children and young adults with treatment-resistant cancer (NCT04730349). It should be noted though that none of these trials is exclusively recruiting non-WNT-/non-SHH or even generally MB-patients, and it remains to be seen if enough patients will be enrolled to allow for subgroup-specific analysis.
 

2.8.2. Cellular Immunotherapy

The success of cellular immunotherapy as a new treatment for hematologic malignancies has sparked intensive research activities that aim at translating these novel therapeutic concepts into the clinic for CNS- and other solid tumors. Currently, two main concepts of cellular immunotherapy are tested in MB patients: chimeric antigen receptor (CAR) T-cells and natural killer (NK) cell therapy []. CAR T-cells are produced by isolating the patient’s own cytotoxic T-cells, which are then equipped with a CAR that can recognize any form of surface marker and subsequently activates the T-cell to mount an immunological attack against the target cell []. It is of crucial importance to choose target antigens that are highly expressed on cancer cells, but not or only negligibly on normal tissue to avoid severe on target/off tumor-toxicity. Currently, several promising targets are investigated for MB-directed CAR T-cell therapy: HER2/Neu, B7-H3 (also called CD276), EPHA2, IL-13Rα2, and PRAME [,,,,,]. HER2/Neu plays an important role as an oncogenic antigen in a number of solid tumors, including breast cancer and glioblastoma, and has also been identified as a possible target for MB-directed cellular therapy both in vitro and in vivo [,,]. Currently, one phase I trial testing HER2/Neu-specific CAR T-cells is recruiting and open to MB patients (NCT03500991), and a recently published interim analysis of the first three enrolled patients (anaplastic astrocytoma, ependymoma) showed no dose-limiting toxicity and presented evidence of immune activation (Table 1) []. B7-H3/CD276 is a pancancer antigen that is strongly expressed by MB []. Similar to HER2/Neu, B7-H3-specific CAR T-cells have shown preclinical activity against non-WNT/non-SHH MB-models both in vitro and in vivo as well as in a number of other (pediatric) cancers, including atypical teratoid/rhabdoid tumor (AT/RT), another aggressive CNS-tumor of early childhood [,,,]. These preclinical findings provide a strong rationale to test B7-H3 targeting CAR T-cells in the clinic, which is currently undertaken in one phase I study that enrolls children with B7-H3 positive CNS-malignancies, including MB (NCT04185038). While HER2/Neu and B7-H3 targeting CARs have already been translated into the clinic, several other targets may be of interest based on preclinical data: one study showed strong expression of EPHA2 and IL-13α2 in human Gr.3 MB and subsequently tested both monovalent EPHA2- and trivalent EPHA2-/HER2/Neu-/IL-13α2-targeting CAR T-cells in a Gr. 3 MB-mouse model, with promising results []. An IL-13α2 directed CAR T-cell trial is recruiting adult patients with leptomeningeal metastases, including MB, and the results may be of interest to inform future trials in pediatric populations (NCT04661384). Furthermore, another study provided evidence in vitro that PRAME might represent a promising target for CAR T-cell therapy in MB []. Additionally, two more phase I CAR-T cell trials are currently open to pediatric patients with CNS-malignancies, including MB. However, the respective target antigens EGFR806 and GD2 have not been tested preclinically in MB patients (NCT03638167 and NCT04099797). Lastly, it should be noted that the delivery of CAR T-cells to the brain poses challenges due to the role of the blood–brain barrier, which could lower the effectiveness of intravenously applied cellular immunotherapies []. To date, the best application route for CAR T-cell in neurooncology has not yet been determined. However, the majority (4/5) of currently running CAR T-cell trials for which MB patients are eligible will use intraventricular/intracavital cell delivery, which circumvents the blood–brain barrier.

An exciting alternative to CAR T-cells is the use of NK cells that may offer certain advantages in comparison, such as lower side effects and increased resistance to immune evasion strategies of the tumor []. Several preclinical studies have shown that in principle, NK cells are able to recognize and eliminate MB cells; however, additional stimulation may be needed to arrive at clinically meaningful cytotoxic activity levels [,,,,]. Interestingly, one study showed a higher and more consistent sensitivity to NK cells in vitro for non-WNT/non-SHH as compared to SHH MB cell lines []. In contrast, another study reported significantly higher expression levels of CD1d, an antigen recognized by NK cells, on SHH as compared to Gr. 4 MB []. Clearly, further studies are needed to arrive at a conclusive answer concerning which MB groups are the most promising target for NK cell therapy. The safety and feasibility of NK cell therapy for pediatric brain cancer has recently been shown by a phase I trial that also enrolled five MB patients, although without reporting molecular diagnoses (NCT02271711) [].

 

2.8.3. Tumor Vaccinations

Cancer vaccine approaches harness the ability of off-the-shelf or patient-specific antigens to induce an antitumoral immune reaction []. Several different strategies have been developed, for instance using antigen pulsed dendritic cells, peptides, and nucleic acid vectors. Tumor vaccines have been studied for a long time; however, several early phase trials have recently shown great potential in adult glioblastoma patients [,,]. One study tested dendritic cells that were pulsed with tumor lysate-derived antigens. As compared to AT/RT and high grade glioma, the five included MB patients showed only modest therapy response, albeit no molecular information is available []. The results of another phase I trial that used RNA-pulsed dendritic cells and also enrolled patients with recurrent MB and glioma are pending (NCT03615404). An additional trial applies another strategy that uses a peptide-based approach (NCT03299309). Lastly, MB patients are currently also eligible for a first-in-pediatrics trial that investigates a long peptide vaccine that targets the apoptosis inhibitor survivin (NCT04978727) (Table 1). Similar to most of the treatment strategies presented so far, none of these trials is restricted to non-WNT/non-SHH MB. Furthermore, so far, no definitive conclusions can be drawn from the reported data concerning differences in the efficacy of vaccination therapies for MB molecular subgroups.

 

2.8.4. Other Immunotherapeutic Approaches

Apart from checkpoint inhibition, increasingly more immunomodulatory approaches are entering the clinical stage. Two studies for a mixed pediatric population with CNS malignancies are currently testing immunostimulatory agents (Table 1): firstly, the antibody APX005M that targets CD40 (NCT03389802) and secondly the immune modulator WP1066, which inhibits the transcription factor STAT3 and therefore interferes with the JAK2/STAT3-pathway (NCT04334863). Additionally, oncolytic viruses, which have recently shown promising results for pediatric high grade glioma, have been proposed as a therapeutic option for MB [,]. Preclinical studies suggest that Gr. 3 MB might be a potential candidate for oncolytic virus therapy, and several different viral vectors have been tested throughout the last decade [,,,,,,,,]. Oncolytic virus studies that are currently recruiting MB patients include the investigation of a modified measles vaccine (NCT02962167), Herpes Simplex Virus (NCT03911388), and one open phase IB trial that will assess possible agonistic effects between mesenchymal allogenic cells and an adenovirus-based virotherapy (NCT04758533) (Table 1). Two additional early phase studies that are open to MB patients are currently active, but not recruiting; these are testing reovirus in combination with Sagramostim and a modified poliovirus (NCT02444546NCT03043391).

2.9. Other Molecular Therapeutic Approaches

In addition to the treatment approaches discussed thus far, several other concepts should be mentioned briefly (Table 1). Firstly, the inhibition of placental growth factor (PGF) has been proposed as a promising strategy across all MB subgroups and was tested in a phase I trial using the monoclonal antibody TB-403 (NCT02748135) []. The results have been presented at the AACR annual meeting 2021 and were encouraging both in terms of safety and efficacy according to the producing company; however, a peer-reviewed publication is not available to date []. Another interesting treatment approach represents the inhibition of DNA damage response pathways using poly (ADP-ribose) polymerase (PARP) inhibitors, a relatively new drug class that has received numerous approvals for breast and ovarian cancer patients with germline BRCA1/2 mutations in the last years []. This could be especially relevant to a subset Gr. 3/4 MB, since these are the molecular groups that are enriched for (germline) BRCA2- and PALB2-mutations [,]. Thus, it will be interesting to see whether non-WNT/non-SHH patients will be included in currently running trials using PARP inhibitors (NCT03233204 and NCT04236414). Lastly, several preclinical studies with different compounds indicate that the concept of sensitizing cancer cells to radiotherapy using small molecules could be potentially interesting for non-WNT/non-SHH MB [,,]

 

(責(zé)任編輯:佳學(xué)基因)
頂一下
(0)
0%
踩一下
(0)
0%
推薦內(nèi)容:
來了,就說兩句!
請(qǐng)自覺遵守互聯(lián)網(wǎng)相關(guān)的政策法規(guī),嚴(yán)禁發(fā)布色情、暴力、反動(dòng)的言論。
評(píng)價(jià):
表情:
用戶名: 驗(yàn)證碼: 點(diǎn)擊我更換圖片

Copyright © 2013-2033 網(wǎng)站由佳學(xué)基因醫(yī)學(xué)技術(shù)(北京)有限公司,湖北佳學(xué)基因醫(yī)學(xué)檢驗(yàn)實(shí)驗(yàn)室有限公司所有 京ICP備16057506號(hào)-1;鄂ICP備2021017120號(hào)-1

設(shè)計(jì)制作 基因解碼基因檢測(cè)信息技術(shù)部