Immunotherapy resistance

免疫治疗耐药
  • 文章类型: Journal Article
    尽管抗程序性细胞死亡-1(抗PD-1)免疫疗法取得了成功,许多癌症患者仍然没有反应,缺乏可靠的预测生物标志物。这里,我们表明嘧啶能受体P2RY6的异常表达在人类癌症中很常见,并导致免疫逃避。在小鼠同基因和人类异种移植肿瘤模型中,P2RY6的异位表达形成免疫抑制性肿瘤微环境(TME),以增强肿瘤生长和对免疫治疗的抵抗力,而P2RY6从高P2RY6表达的肿瘤中缺失会使TME发炎以抑制肿瘤生长。作为G蛋白偶联受体,P2RY6激活Gq/磷脂酶C-β信号并刺激前列腺素E2的合成,前列腺素E2是TME中免疫抑制的关键介质。与P2RY6在肿瘤中的重要作用相反,从小鼠中全局缺失P2ry6不会损害生存能力。因此,我们的研究将P2RY6作为肿瘤固有P2RY6高表达患者的精确免疫疗法靶标。
    Despite the success of anti-programmed cell death-1 (anti-PD-1) immunotherapy, many cancer patients remain unresponsive, and reliable predictive biomarkers are lacking. Here, we show that aberrant expression of the pyrimidinergic receptor P2RY6 is frequent in human cancers and causes immune evasion. In mouse syngeneic and human xenograft tumor models, ectopic expression of P2RY6 shapes an immunosuppressive tumor microenvironment (TME) to enhance tumor growth and resistance to immunotherapy, whereas deletion of P2RY6 from tumors with high P2RY6 expression inflames the TME to inhibit tumor growth. As a G protein-coupled receptor, P2RY6 activates Gq/phospholipase C-β signaling and stimulates the synthesis of prostaglandin E2, which is a key mediator of immunosuppression in the TME. In contrast to the essential role of P2RY6 in tumors, global deletion of P2ry6 from mice does not compromise viability. Our study thus nominates P2RY6 as a precision immunotherapy target for patients with high tumor-intrinsic P2RY6 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    微卫星不稳定性高(MSI-H)结直肠癌(CRC)对免疫检查点抑制剂(ICI)的治疗反应确实令人惊讶;然而,获得性耐药性的出现对这些患者的生存构成了更大的威胁。在这里,MSI-HCRC样本的生物信息学分析显示,Wnt信号通路是获得性免疫再激活的一个有希望的目标,而随后的分析和生化测试证实了Wnt过度活跃的CRC细胞倾向于与人血清白蛋白(HSA)一起进行大胞吞作用。这些发现激励我们开发一种工程化的HSA,其不仅具有特异性靶向癌细胞的能力,而且还有效地抑制这些恶性细胞内的Wnt/β-连环蛋白级联。为了实现这个目标,对已报道的Wnt小分子抑制剂进行了全面筛选,以评估其与HSA的亲和力,并且发现鼠尾草酸(CA)表现出最高的亲和力,同时揭示多个结合位点。进一步的调查显示,CAHSA能够将HSA设计成球形和尺寸可调的纳米结构,称为eHSA(工程HSA颗粒)。这证明了优化的巨细胞增多依赖性细胞内化。如预期,eHSA有效抑制Wnt信号通路并在体内重新激活获得性免疫应答。此外,在MSI-HCRC的皮下和原位小鼠同源移植模型中,eHSA成功恢复了对抗PD1抗癌作用的敏感性,以及人源化hu-PBMC患者来源的MSI-HCRC原位异种移植(PDOX)小鼠模型,同时保持良好的安全性。这种临床上可行的免疫再激活策略的集体实施不仅能够提供用于CRC治疗的Wnt抑制剂。但也可以作为精确医学指导的纳米药物开发的示例性演示,该药物开发可以有效地利用病理状态下的特定细胞适应症。
    The therapeutic response of microsatellite instability-high (MSI-H) colorectal cancer (CRC) to immune checkpoint inhibitors (ICI) is indeed surprising; however, the emergence of acquired resistance poses an even greater threat to the survival of these patients. Herein, bioinformatics analysis of MSI-H CRC samples revealed that Wnt signaling pathway represents a promising target for acquired immune reactivation, while subsequent analysis and biochemical testing substantiated the inclination of Wnt-hyperactive CRC cells to engage in macropinocytosis with human serum albumin (HSA). These findings have inspired us to develop an engineered HSA that not only possesses the ability to specifically target cancer cells but also effectively suppresses the Wnt/β-catenin cascade within these malignant cells. In pursuit of this objective, a comprehensive screening of reported Wnt small-molecule inhibitors was conducted to evaluate their affinity with HSA, and it was discovered that Carnosic acid (CA) exhibited the highest affinity while simultaneously revealing multiple binding sites. Further investigation revealed that CA HSA the capability to engineer HSA into spherical and size-tunable nanostructures known as eHSA (Engineering HSA particle), which demonstrated optimized macropinocytosis-dependent cellular internalization. As anticipated, eHSA effectively suppressed the Wnt signaling pathway and reactivated the acquired immune response in vivo. Furthermore, eHSA successfully restored sensitivity to Anti-PD1\'s anticancer effects in both subcutaneous and orthotopic mouse homograft models of MSI-H CRC, as well as a humanized hu-PBMC patient-derived orthotopic xenograft (PDOX) mouse model of MSI-H CRC, all while maintaining a favorable safety profile. The collective implementation of this clinically viable immune reactivation strategy not only enables the delivery of Wnt inhibitors for CRC therapy, but also serves as an exemplary demonstration of precision-medicine-guided nanopharmaceutical development that effectively harnesses specific cellular indications in pathological states.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    抗程序性死亡1/程序性死亡配体1(抗PD-1/PD-L1)抗体通过克服肿瘤细胞免疫逃避和逆转T细胞耗竭而发挥显著的抗肿瘤作用。然而,耐药性的出现导致大多数患者对这些免疫检查点抑制剂(ICIs)的反应较差.研究表明,T细胞浸润不足,缺乏PD-1表达,缺乏干扰素信号,肿瘤抗原呈递的丧失,脂质代谢异常均被认为与免疫治疗耐药密切相关。为了解决肿瘤免疫治疗中的耐药性,许多研究集中在开发联合治疗策略上。目前,抗PD-1/PD-L1抗体等ICI联合化疗和靶向治疗已被批准用于临床治疗。在这次审查中,我们从肿瘤微环境的角度分析抗PD-1/PD-L1治疗的耐药机制,肠道菌群,表观遗传调控,和共抑制性免疫检查点受体。我们还讨论了各种有前途的联合治疗策略,以解决抗PD-1/PD-L1药物的耐药性,包括将这些疗法与传统中药相结合,非编码RNA,靶向治疗,其他ICIs,和个性化的癌症疫苗。此外,我们关注预测抗PD-1/PD-L1治疗耐药以及联合治疗疗效的生物标志物.最后,我们提出了通过使用生物标志物系统的个性化组合策略进一步扩大免疫治疗应用的方法.
    Anti-programmed death 1/programmed death ligand 1 (anti-PD-1/PD-L1) antibodies exert significant antitumor effects by overcoming tumor cell immune evasion and reversing T-cell exhaustion. However, the emergence of drug resistance causes most patients to respond poorly to these immune checkpoint inhibitors (ICIs). Studies have shown that insufficient T-cell infiltration, lack of PD-1 expression, deficient interferon signaling, loss of tumor antigen presentation, and abnormal lipid metabolism are all considered to be closely associated with immunotherapy resistance. To address drug resistance in tumor immunotherapy, a lot of research has concentrated on developing combination therapy strategies. Currently, ICIs such as anti-PD-1 /PD-L1 antibody combined with chemotherapy and targeted therapy have been approved for clinical treatment. In this review, we analyze the mechanisms of resistance to anti-PD-1/PD-L1 therapy in terms of the tumor microenvironment, gut microbiota, epigenetic regulation, and co-inhibitory immune checkpoint receptors. We also discuss various promising combination therapeutic strategies to address resistance to anti-PD-1/PD-L1 drugs, including combining these therapies with traditional Chinese medicine, non-coding RNAs, targeted therapy, other ICIs, and personalized cancer vaccines. Moreover, we focus on biomarkers that predict resistance to anti-PD-1/PD-L1 therapy as well as combination therapy efficacy. Finally, we suggest ways to further expand the application of immunotherapy through personalized combination strategies using biomarker systems.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    免疫疗法彻底改变了某些癌症的治疗方法,但是在克服免疫疗法耐药性方面仍然存在挑战。研究表明,肿瘤微环境的代谢调节可以增强抗肿瘤免疫力。这里,我们讨论了代谢修饰剂与免疫疗法联合应用的近期临床前和临床证据.虽然这种组合很有希望,必须解决几个关键领域,其中包括确定代谢调节剂对免疫细胞代谢的影响,治疗功效的推定生物标志物,修饰剂对具有代谢异质性的肿瘤的功效,以及由于肿瘤对替代代谢途径的依赖而导致的耐药性的潜在发展。我们提出了这些问题的解决方案,并认为评估这些参数对于考虑代谢调节剂在使肿瘤对免疫疗法敏感方面的潜力至关重要。
    Immunotherapies have revolutionized the treatment of certain cancers, but challenges remain in overcoming immunotherapy resistance. Research shows that metabolic modulation of the tumor microenvironment can enhance antitumor immunity. Here, we discuss recent preclinical and clinical evidence for the efficacy of combining metabolic modifiers with immunotherapies. While this combination holds great promise, a few key areas must be addressed, which include identifying the effects of metabolic modifiers on immune cell metabolism, the putative biomarkers of therapeutic efficacy, the efficacy of modifiers on tumors harboring metabolic heterogeneity, and the potential development of resistance due to tumor reliance on alternative metabolic pathways. We propose solutions to these problems and posit that assessing these parameters is crucial for considering the potential of metabolic modifiers in sensitizing tumors to immunotherapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    通过以疫苗为基础的癌症免疫疗法,已经开始在实际临床应用中取得突破,它使用人体的免疫系统,体液和细胞,攻击恶性细胞和对抗疾病。然而,传统的疫苗方法仍然面临着引发有效的抗原特异性免疫反应的多重挑战,导致免疫疗法耐药。近年来,通过结合各种生物实体的天然结构,仿生纳米疫苗已成为传统疫苗方法的有希望的替代品,如细胞,病毒,和细菌。仿生纳米疫苗提供靶向抗原呈递细胞(APC)递送的好处,改进的抗原/佐剂负载,和生物相容性,从而提高免疫治疗的敏感性。本文综述了几种仿生纳米疫苗在抗肿瘤免疫反应中的作用,包括细胞膜包覆的纳米疫苗,基于蛋白质的自组装纳米疫苗,基于细胞外囊泡的纳米疫苗,天然配体修饰的纳米疫苗,基于人工抗原呈递细胞的纳米疫苗和基于脂质体的纳米疫苗。我们还讨论了与新兴的仿生纳米疫苗平台的临床翻译相关的观点和挑战,这些平台用于使癌细胞对免疫疗法敏感。
    Breakthroughs in actual clinical applications have begun through vaccine-based cancer immunotherapy, which uses the body\'s immune system, both humoral and cellular, to attack malignant cells and fight diseases. However, conventional vaccine approaches still face multiple challenges eliciting effective antigen-specific immune responses, resulting in immunotherapy resistance. In recent years, biomimetic nanovaccines have emerged as a promising alternative to conventional vaccine approaches by incorporating the natural structure of various biological entities, such as cells, viruses, and bacteria. Biomimetic nanovaccines offer the benefit of targeted antigen-presenting cell (APC) delivery, improved antigen/adjuvant loading, and biocompatibility, thereby improving the sensitivity of immunotherapy. This review presents a comprehensive overview of several kinds of biomimetic nanovaccines in anticancer immune response, including cell membrane-coated nanovaccines, self-assembling protein-based nanovaccines, extracellular vesicle-based nanovaccines, natural ligand-modified nanovaccines, artificial antigen-presenting cells-based nanovaccines and liposome-based nanovaccines. We also discuss the perspectives and challenges associated with the clinical translation of emerging biomimetic nanovaccine platforms for sensitizing cancer cells to immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:西妥昔单抗抑制表皮生长因子受体(EGFR)是头颈部鳞状细胞癌(HNSCC)的标准治疗方法。受体酪氨酸激酶AXL的激活,MET和VEGFR可以介导对西妥昔单抗的抗性。卡博替尼,多激酶抑制剂(MKI)靶向AXL/MET/VEGFR,在HNSCC的临床前模型中已证明具有抗肿瘤活性。这项研究者启动的I期试验评估了西妥昔单抗联合卡博替尼在复发/转移性(R/M)HNSCC患者中的安全性和有效性。
    方法:患者在28天的周期内每天同时接受西妥昔单抗和卡博替尼。使用3+3剂量递增设计,主要终点是确定卡博替尼的最大耐受剂量(MTD).次要终点包括总体反应率(ORR),疾病控制率(DCR),无进展生存期(PFS),和总生存期(OS)结果:在20名患者中,大多数人在免疫检查点抑制剂(95%)之前有疾病进展,铂类化疗(95%),西妥昔单抗(80%)。没有记录剂量限制性毒性,并且卡博替尼的MTD确定为60mg。65%的患者发生≥3级不良事件(n=13)。ORR为20%,有4个部分响应(PR)。在西妥昔单抗初治患者中观察到两个PR(n=4),在这个亚组中ORR为50%。在总人口中,DCR为75%,中位PFS为3.4个月,中位OS为8.1个月.
    结论:西妥昔单抗联合卡博替尼在重度治疗的R/MHNSCC患者中表现出可控的毒性特征和初步疗效。西妥昔单抗与针对AXL/MET/VEGFR轴的MKI的组合值得进一步研究,包括西妥昔单抗初治患者。
    OBJECTIVE: Epidermal growth factor receptor (EGFR) inhibition with cetuximab is a standard treatment for head and neck squamous cell carcinoma (HNSCC). Activation of the receptor tyrosine kinases AXL, MET and VEGFR can mediate resistance to cetuximab. Cabozantinib, a multikinase inhibitor (MKI) targeting AXL/MET/VEGFR, has demonstrated antitumor activity in preclinical models of HNSCC. This investigator- initiated phase I trial evaluated the safety and efficacy of cetuximab plus cabozantinib in patients with recurrent/metastatic (R/M) HNSCC.
    METHODS: Patients received cetuximab concurrently with cabozantinib daily on a 28-day cycle. Using a 3 + 3 dose-escalation design, the primary endpoint was to determine the maximally tolerated dose (MTD) of cabozantinib. Secondary endpoints included overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) RESULTS: Among the 20 patients enrolled, most had prior disease progression on immune checkpoint inhibitors (95 %), platinum-based chemotherapy (95 %), and cetuximab (80 %). No dose-limiting toxicities were recorded and the MTD for cabozantinib was established to be 60 mg. Grade ≥ 3 adverse events occurred in 65 % of patients (n = 13). ORR was 20 %, with 4 partial responses (PRs). Two PRs were observed in cetuximab-naïve patients (n = 4), with an ORR of 50 % in this subgroup. In the overall population, DCR was 75 %, median PFS was 3.4 months and median OS was 8.1 months.
    CONCLUSIONS: Cetuximab plus cabozantinib demonstrated a manageable toxicity profile and preliminary efficacy in patients with heavily treated R/M HNSCC. The combination of cetuximab with MKIs targeting the AXL/MET/VEGFR axis warrants further investigation, including in cetuximab-naïve patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    新型免疫疗法的引入极大地改变了泌尿生殖系统(GU)癌症的治疗前景,甚至成为某些环境下的护理标准。一种这样的免疫疗法,免疫检查点抑制剂(ICIs),如nivolumab,ipilimumab,pembrolizumab,阿替珠单抗通过干扰信号通路发挥关键作用,这些信号通路限制了免疫系统对抗肿瘤细胞的能力。尽管这些治疗方法产生了深远的影响,并非所有肿瘤都有反应。最近的研究工作集中在了解癌细胞如何设法逃避免疫反应,并确定对免疫疗法耐药背后的可能机制。作为回应,ICI正在与其他治疗相结合,以减少耐药性并通过多种细胞途径攻击癌细胞。此外,小说,目前正在研究有针对性的策略,以开发克服耐药性和治疗失败的创新方法。本文对目前文献中描述的GU癌症中免疫疗法耐药的机制进行了全面概述。它探索了确定遗传标记的研究,细胞因子,和可以预测对免疫疗法的抗性或反应的蛋白质。此外,我们回顾了当前克服这种阻力的努力,其中包括联合ICIs和序贯疗法,对宿主免疫谱的新见解,和新的靶向疗法。各种联合免疫疗法和化疗的方法,靶向治疗,疫苗,为了更有效地克服对免疫疗法的抗性,已经研究了辐射。虽然这些联合疗法在临床试验中都显示出一定的疗效,对免疫系统作用的更深入的了解强调了新型靶向疗法作为当前研究中一个特别有前景的领域的潜力。目前,几个目标代理正在开发中,以及与免疫疗法抗性有关的关键免疫介质的鉴定。需要进一步的研究来确定反应的预测因素。
    免疫治疗改变了许多癌症类型的治疗前景。包括泌尿生殖系统恶性肿瘤,如肾癌和膀胱癌。然而,不是所有的病人或肿瘤类型,比如前列腺癌,对这种治疗的反应。了解免疫疗法抵抗的机制对于制定克服这些挑战的策略至关重要。主要阻力,在治疗开始时存在,可能是由于遗传异常或免疫系统失调。这些因素改变宿主细胞和癌细胞之间的相互作用。由于生长因子水平的动态变化,在治疗过程中会产生适应性抵抗,细胞因子,和肿瘤微环境(TME)。获得性抗性主要发生在遗传和翻译水平,涉及关键人类白细胞抗原(HLA)分子的下调和对突变修复的干扰。未来的治疗方法可能侧重于患者的详细遗传分析,以指导治疗选择,并使用免疫谱监测来帮助评估反应性。同时开发新的靶向疗法和ICIs。需要进一步的研究来确定对ICI反应的预测因素。
    The introduction of novel immunotherapies has significantly transformed the treatment landscape of genitourinary (GU) cancers, even becoming the standard of care in some settings. One such type of immunotherapy, immune checkpoint inhibitors (ICIs) like nivolumab, ipilimumab, pembrolizumab, and atezolizumab play a pivotal role by disturbing signaling pathways that limit the immune system\'s ability to fight tumor cells. Despite the profound impact of these treatments, not all tumors are responsive. Recent research efforts have been focused on understanding how cancer cells manage to evade the immune response and identifying the possible mechanisms behind resistance to immunotherapy. In response, ICIs are being combined with other treatments to reduce resistance and attack cancer cells through multiple cellular pathways. Additionally, novel, targeted strategies are currently being investigated to develop innovative methods of overcoming resistance and treatment failure. This article presents a comprehensive overview of the mechanisms of immunotherapy resistance in GU cancers as currently described in the literature. It explores studies that have identified genetic markers, cytokines, and proteins that may predict resistance or response to immunotherapy. Additionally, we review current efforts to overcome this resistance, which include combination ICIs and sequential therapies, novel insights into the host immune profile, and new targeted therapies. Various approaches that combine immunotherapy with chemotherapy, targeted therapy, vaccines, and radiation have been studied in an effort to more effectively overcome resistance to immunotherapy. While each of these combination therapies has shown some efficacy in clinical trials, a deeper understanding of the immune system\'s role underscores the potential of novel targeted therapies as a particularly promising area of current research. Currently, several targeted agents are in development, along with the identification of key immune mediators involved in immunotherapy resistance. Further research is necessary to identify predictors of response.
    Immunotherapy has transformed the treatment landscape for many cancer types, including genitourinary malignancies such as renal and bladder cancers.However, not all patients or tumor types, such as prostate cancer, respond to this type of treatment.Understanding the mechanisms of immunotherapy resistance is critical for developing strategies to overcome these challenges.Primary resistance, which is present at the onset of treatment, can bedue to genetic abnormalities or immune system dysregulation. These factors alter the interactions between host cells and cancer cells.Adaptive resistance develops during therapy due to dynamic changes in the levels of growth factors, cytokines, and the tumor microenvironment (TME).Acquired resistance mainly occurs at the genetic and translational levels, involving the downregulation of critical human leukocyte antigen (HLA) molecules and interference with mutational repair.Future therapies may focus on detailed genetic profiling of patients to guide treatment selection and on the use of immune profile monitoring to assist in assessing responsiveness, alongside developing novel targeted therapies and ICIs.Further research is needed to identify predictors of response to ICIs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    免疫疗法,尤其是免疫检查点抑制剂,在过去的十年里彻底改变了临床实践。然而,对免疫疗法的原发性和继发性耐药性在患有不同类型癌症的患者中很常见.众所周知,肿瘤细胞可以通过代谢重编程促进免疫抑制微环境的形成,还有乳酸,糖酵解的代谢产物,是一个重要的贡献者。SLC16A3(也称为MCT4)是介导乳酸流出的转运蛋白。在这项研究中,我们研究了糖酵解在免疫治疗抵抗中的作用,旨在通过Slc16a3抑制改善免疫治疗效果.生物信息学分析显示,糖酵解相关基因的表达与CD8T细胞浸润减少和髓源性抑制细胞(MDSC)富集增加相关。我们发现,肿瘤细胞中的高糖酵解活性会不利地影响抗肿瘤免疫反应以及免疫疗法和放射疗法的功效。作为乳酸的转运蛋白,SLC16A3在糖酵解B16-F10(RRID:CVCL_0159)细胞中高表达,以及人类非小细胞肺癌。我们验证了Slc16a3在肿瘤细胞中的表达与抗PD-1效率呈负相关。Slc16a3在肿瘤细胞中的过表达促进乳酸的产生和流出,并通过抑制CD8+T细胞功能降低肿瘤对抗PD-1抑制剂的反应。Slc16a3的遗传和药理抑制显着降低了肿瘤细胞中的糖酵解活性和乳酸的产生,并改善了免疫抑制肿瘤微环境(TME),导致通过抗PD-1阻断增强的抗肿瘤作用。因此,我们的研究表明,肿瘤细胞固有的SLC16A3可能是逆转肿瘤对免疫疗法耐药性的潜在靶标。
    Immunotherapy, especially immune checkpoint inhibitors, has revolutionized clinical practice within the last decade. However, primary and secondary resistance to immunotherapy is common in patients with diverse types of cancer. It is well-acknowledged that tumor cells can facilitate the formation of immunosuppressive microenvironments via metabolism reprogramming, and lactic acid, the metabolite of glycolysis, is a significant contributor. SLC16A3 (also named as MCT4) is a transporter mediating lactic acid efflux. In this study, we investigated the role of glycolysis in immunotherapy resistance and aimed to improve the immunotherapy effects via Slc16a3 inhibition. Bioinformatical analysis revealed that the expression of glycolysis-related genes correlated with less CD8+ T cell infiltration and increased myeloid-derived suppressor cells (MDSC) enrichment. We found that high glycolytic activity in tumor cells adversely affected the antitumor immune responses and efficacy of immunotherapy and radiotherapy. As the transporter of lactic acid, SLC16A3 is highly expressed in glycolytic B16-F10 (RRID: CVCL_0159) cells, as well as human non-small cell lung carcinoma. We validated that Slc16a3 expression in tumor cells negatively correlated with anti-PD-1 efficiency. Overexpression of Slc16a3 in tumor cells promoted lactic acid production and efflux, and reduced tumor response to anti-PD-1 inhibitors by inhibiting CD8+ T cell function. Genetic and pharmacological inhibition of Slc16a3 dramatically reduced the glycolytic activity and lactic acid production in tumor cells, and ameliorated the immunosuppressive tumor microenvironments (TMEs), leading to boosted antitumor effects via anti-PD-1 blockade. Our study therefore demonstrates that tumor cell-intrinsic SLC16A3 may be a potential target to reverse tumor resistance to immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号