首页 > 最新文献

Molecular Cancer Therapeutics最新文献

英文 中文
Anti-Claudin-2 Antibody-Drug Conjugates for the Treatment of Colorectal Cancer Liver Metastasis. 用于治疗结直肠癌肝转移的抗 Claudin-2 抗体-药物共轭物。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1535-7163.MCT-23-0393
Sébastien Tabariès, Alma Robert, Anne Marcil, Binbing Ling, Mauro Acchione, Julie Lippens, Martine Pagé, Annie Fortin, Luc Meury, Mathieu Coutu, Matthew G Annis, Charlotte Girondel, Julie Navarre, Maria Jaramillo, Anna N Moraitis, Peter M Siegel

We have previously demonstrated that Claudin-2 is required for colorectal cancer (CRC) liver metastasis. The expression of Claudin-2 in primary CRC is associated with poor survival and highly expressed in liver metastases. Claudin-2 also promotes breast cancer liver metastasis by enabling seeding and cancer cell survival. These observations support Claudin-2 as a potential therapeutic target for managing patients with liver metastases. Antibody-drug conjugates (ADC) are promising antitumor therapeutics, which combine the specific targeting ability of monoclonal antibodies with the potent cell killing activity of cytotoxic drugs. Herein, we report the generation of 28 anti-Claudin-2 antibodies for which the binding specificities, cross-reactivity with claudin family members, and cross-species reactivity were assessed by flow cytometry analysis. Multiple drug conjugates were tested, and PNU was selected for conjugation with anti-Claudin-2 antibodies binding either extracellular loop 1 or 2. Anti-Claudin-2 ADCs were efficiently internalized and were effective at killing Claudin-2-expressing CRC cancer cells in vitro. Importantly, PNU-conjugated-anti-Claudin-2 ADCs impaired the development of replacement-type CRC liver metastases in vivo, using established CRC cell lines and patient-derived xenograft (PDX) models of CRC liver metastases. Results suggest that the development of ADCs targeting Claudin-2 is a promising therapeutic strategy for managing patients with CRC liver-metastatic disease who present replacement-type liver metastases.

我们已经证明,Claudin-2 是结直肠癌(CRC)肝转移的必要条件。Claudin-2在原发性结直肠癌中的表达与生存率低有关,并且在肝转移中高度表达。Claudin-2还通过促进播种和癌细胞存活来促进乳腺癌的肝转移。这些观察结果支持将 Claudin-2 作为治疗肝转移患者的潜在治疗靶点。抗体药物共轭物(ADCs)是一种前景广阔的抗肿瘤疗法,它结合了单克隆抗体的特异性靶向能力和细胞毒性药物的强大细胞杀伤活性。在此,我们报告了 28 种抗 Claudin-2 抗体的生成情况,并通过流式细胞术分析评估了这些抗体的结合特异性、与 Claudin 家族成员的交叉反应性以及跨物种反应性。测试了多种药物共轭物,最后选择了 PNU 与结合细胞外环 1 或细胞外环 2 的抗 Claudin-2 抗体共轭。抗Claudin-2 ADC能在体外有效内化并杀死表达Claudin-2的CRC癌细胞。重要的是,利用已建立的 CRC 细胞系和 CRC 肝转移患者衍生异种移植(PDX)模型,PNU 结合物-抗 Claudin-2 ADCs 在体内可抑制替代型 CRC 肝转移灶的发展。我们的研究结果表明,开发以Claudin-2为靶点的ADCs是治疗出现替代型肝转移的CRC肝转移患者的一种很有前景的治疗策略。
{"title":"Anti-Claudin-2 Antibody-Drug Conjugates for the Treatment of Colorectal Cancer Liver Metastasis.","authors":"Sébastien Tabariès, Alma Robert, Anne Marcil, Binbing Ling, Mauro Acchione, Julie Lippens, Martine Pagé, Annie Fortin, Luc Meury, Mathieu Coutu, Matthew G Annis, Charlotte Girondel, Julie Navarre, Maria Jaramillo, Anna N Moraitis, Peter M Siegel","doi":"10.1158/1535-7163.MCT-23-0393","DOIUrl":"10.1158/1535-7163.MCT-23-0393","url":null,"abstract":"<p><p>We have previously demonstrated that Claudin-2 is required for colorectal cancer (CRC) liver metastasis. The expression of Claudin-2 in primary CRC is associated with poor survival and highly expressed in liver metastases. Claudin-2 also promotes breast cancer liver metastasis by enabling seeding and cancer cell survival. These observations support Claudin-2 as a potential therapeutic target for managing patients with liver metastases. Antibody-drug conjugates (ADC) are promising antitumor therapeutics, which combine the specific targeting ability of monoclonal antibodies with the potent cell killing activity of cytotoxic drugs. Herein, we report the generation of 28 anti-Claudin-2 antibodies for which the binding specificities, cross-reactivity with claudin family members, and cross-species reactivity were assessed by flow cytometry analysis. Multiple drug conjugates were tested, and PNU was selected for conjugation with anti-Claudin-2 antibodies binding either extracellular loop 1 or 2. Anti-Claudin-2 ADCs were efficiently internalized and were effective at killing Claudin-2-expressing CRC cancer cells in vitro. Importantly, PNU-conjugated-anti-Claudin-2 ADCs impaired the development of replacement-type CRC liver metastases in vivo, using established CRC cell lines and patient-derived xenograft (PDX) models of CRC liver metastases. Results suggest that the development of ADCs targeting Claudin-2 is a promising therapeutic strategy for managing patients with CRC liver-metastatic disease who present replacement-type liver metastases.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141432331","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
AKT Inhibition Sensitizes to Polo-Like Kinase 1 Inhibitor Onvansertib in Prostate Cancer. AKT 抑制使前列腺癌患者对 Polo-Like Kinase 1 抑制剂 Onvansertib 敏感
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1535-7163.MCT-23-0933
Mannan Nouri, Andreas Varkaris, Maya Ridinger, Susan L Dalrymple, Christopher M Dennehy, John T Isaacs, David J Einstein, W N Brennen, Steven P Balk

Polo-like kinase 1 (PLK1) inhibitors have had limited antitumor efficacy as single agents, and focus of current efforts is on combination therapies. We initially confirmed that the PLK1-specific inhibitor onvansertib (ONV) could enhance responses to a PARP inhibitor (olaparib) in prostate cancer xenografts. To identify more effective combinations, we screened a library of bioactive compounds for efficacy in combination with ONV in LNCaP prostate cancer cells, which identified a series of compounds including multiple AKT inhibitors. We confirmed in vitro synergy between ONV and the AKT inhibitor ipatasertib (IPA) and found that the combination increased apoptosis. Mechanistic studies showed that ONV increased expression of the antiapoptotic protein SURVIVIN and that this was mitigated by IPA. Studies in three PTEN-deficient prostate cancer xenograft models showed that cotreatment with IPA and ONV led to significant tumor growth inhibition compared with monotherapies. Together, these in vitro and in vivo studies demonstrate that the efficacy of PLK1 antagonists can be enhanced by PARP or AKT inhibition and support further development of these combination therapies.

Polo-like激酶1(PLK1)抑制剂作为单药的抗肿瘤疗效有限,目前的工作重点是联合疗法。我们初步证实,PLK1特异性抑制剂onvansertib(ONV)能增强前列腺癌异种移植物对PARP抑制剂(奥拉帕尼)的反应。为了确定更有效的组合,我们筛选了一个生物活性化合物库,以确定与 ONV 在 LNCaP 前列腺癌细胞中的联合疗效,结果发现了一系列化合物,包括多种 AKT 抑制剂。我们证实了 ONV 与 AKT 抑制剂 ipatasertib(IPA)之间的体外协同作用,并发现这种组合能增加细胞凋亡。机理研究表明,ONV 增加了抗凋亡蛋白 SURVIVIN 的表达,而 IPA 则减轻了这种情况。对三种缺乏 PTEN 的前列腺癌异种移植模型的研究表明,与单一疗法相比,IPA 和 ONV 联合治疗可显著抑制肿瘤生长。这些体外和体内研究共同证明,PARP 或 AKT 抑制可增强 PLK1 拮抗剂的疗效,并支持进一步开发这些联合疗法。
{"title":"AKT Inhibition Sensitizes to Polo-Like Kinase 1 Inhibitor Onvansertib in Prostate Cancer.","authors":"Mannan Nouri, Andreas Varkaris, Maya Ridinger, Susan L Dalrymple, Christopher M Dennehy, John T Isaacs, David J Einstein, W N Brennen, Steven P Balk","doi":"10.1158/1535-7163.MCT-23-0933","DOIUrl":"10.1158/1535-7163.MCT-23-0933","url":null,"abstract":"<p><p>Polo-like kinase 1 (PLK1) inhibitors have had limited antitumor efficacy as single agents, and focus of current efforts is on combination therapies. We initially confirmed that the PLK1-specific inhibitor onvansertib (ONV) could enhance responses to a PARP inhibitor (olaparib) in prostate cancer xenografts. To identify more effective combinations, we screened a library of bioactive compounds for efficacy in combination with ONV in LNCaP prostate cancer cells, which identified a series of compounds including multiple AKT inhibitors. We confirmed in vitro synergy between ONV and the AKT inhibitor ipatasertib (IPA) and found that the combination increased apoptosis. Mechanistic studies showed that ONV increased expression of the antiapoptotic protein SURVIVIN and that this was mitigated by IPA. Studies in three PTEN-deficient prostate cancer xenograft models showed that cotreatment with IPA and ONV led to significant tumor growth inhibition compared with monotherapies. Together, these in vitro and in vivo studies demonstrate that the efficacy of PLK1 antagonists can be enhanced by PARP or AKT inhibition and support further development of these combination therapies.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11444904/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141419899","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Characterization of AB598, a CD39 Enzymatic Inhibitory Antibody for the Treatment of Solid Tumors. 用于治疗实体瘤的 CD39 酶抑制抗体 AB598 的特性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1535-7163.MCT-23-0865
Amy E Anderson, Kaustubh Parashar, Ke Jin, Julie Clor, Carlo E Stagnaro, Urvi Vani, Jaskirat Singh, Ada Chen, Yihong Guan, Priyanka Talukdar, Pavithra Sathishkumar, Damie J Juat, Hema Singh, Ritu Kushwaha, Xiaoning Zhao, Angelo Kaplan, Lisa Seitz, Matthew J Walters, Ester Fernandez-Salas, Nigel P C Walker, Christine E Bowman

AB598 is a CD39 inhibitory antibody being pursued for the treatment of solid tumors in combination with chemotherapy and immunotherapy. CD39 metabolizes extracellular adenosine triphosphate (eATP), an alarmin capable of promoting antitumor immune responses, into adenosine, an immuno-inhibitory metabolite. By inhibiting CD39, the consumption of eATP is reduced, resulting in a proinflammatory milieu in which eATP can activate myeloid cells to promote antitumor immunity. The preclinical characterization of AB598 provides a mechanistic rationale for combining AB598 with chemotherapy in the clinic. Chemotherapy can induce ATP release from tumor cells and, when preserved by AB598, both chemotherapy-induced eATP and exogenously added ATP promote the function of monocyte-derived dendritic cells via P2Y11 signaling. Inhibition of CD39 in the presence of ATP can promote inflammasome activation in in vitro-derived macrophages, an effect mediated by P2X7. In a MOLP8 murine xenograft model, AB598 results in full inhibition of intratumoral CD39 enzymatic activity, an increase in intratumoral ATP, a decrease of extracellular CD39 on tumor cells, and ultimately, control of tumor growth. In cynomolgus monkeys, systemic dosing of AB598 results in effective enzymatic inhibition in tissues, full peripheral and tissue target engagement, and a reduction in cell surface CD39 both in tissues and in the periphery. Taken together, these data support a promising therapeutic strategy of harnessing the eATP generated by standard-of-care chemotherapies to prime the tumor microenvironment for a productive antitumor immune response.

AB598 是一种 CD39 抑制抗体,目前正与化疗和免疫疗法联合用于治疗实体瘤。CD39 能将细胞外 ATP(eATP)(一种能促进抗肿瘤免疫反应的刺激物)代谢为腺苷(一种免疫抑制代谢物)。通过抑制 CD39,可以减少 eATP 的消耗,从而形成一种促炎环境,在这种环境中,eATP 可以激活髓系细胞,促进抗肿瘤免疫。AB598 的临床前特性为在临床中将 AB598 与化疗相结合提供了机理依据。化疗可诱导肿瘤细胞释放 ATP,在 AB598 的保护下,化疗诱导的 eATP 和外源添加的 ATP 都能通过 P2Y11 信号促进单核细胞衍生树突状细胞的功能。在存在 ATP 的情况下抑制 CD39 可促进体外衍生巨噬细胞中炎性小体的活化,这种效应由 P2X7 介导。在 MOLP8 小鼠异种移植模型中,AB598 可完全抑制瘤内酶活性,增加瘤内 ATP,减少肿瘤细胞上的细胞外 CD39,最终控制肿瘤生长。在猴体内,全身给药的 AB598 能有效抑制组织内的酶活性,使外周和组织靶点充分参与,并减少组织和外周的细胞表面 CD39。综上所述,这些数据支持一种前景广阔的治疗策略,即利用标准化疗药物产生的eATP为肿瘤微环境提供能量,从而产生富有成效的抗肿瘤免疫反应。
{"title":"Characterization of AB598, a CD39 Enzymatic Inhibitory Antibody for the Treatment of Solid Tumors.","authors":"Amy E Anderson, Kaustubh Parashar, Ke Jin, Julie Clor, Carlo E Stagnaro, Urvi Vani, Jaskirat Singh, Ada Chen, Yihong Guan, Priyanka Talukdar, Pavithra Sathishkumar, Damie J Juat, Hema Singh, Ritu Kushwaha, Xiaoning Zhao, Angelo Kaplan, Lisa Seitz, Matthew J Walters, Ester Fernandez-Salas, Nigel P C Walker, Christine E Bowman","doi":"10.1158/1535-7163.MCT-23-0865","DOIUrl":"10.1158/1535-7163.MCT-23-0865","url":null,"abstract":"<p><p>AB598 is a CD39 inhibitory antibody being pursued for the treatment of solid tumors in combination with chemotherapy and immunotherapy. CD39 metabolizes extracellular adenosine triphosphate (eATP), an alarmin capable of promoting antitumor immune responses, into adenosine, an immuno-inhibitory metabolite. By inhibiting CD39, the consumption of eATP is reduced, resulting in a proinflammatory milieu in which eATP can activate myeloid cells to promote antitumor immunity. The preclinical characterization of AB598 provides a mechanistic rationale for combining AB598 with chemotherapy in the clinic. Chemotherapy can induce ATP release from tumor cells and, when preserved by AB598, both chemotherapy-induced eATP and exogenously added ATP promote the function of monocyte-derived dendritic cells via P2Y11 signaling. Inhibition of CD39 in the presence of ATP can promote inflammasome activation in in vitro-derived macrophages, an effect mediated by P2X7. In a MOLP8 murine xenograft model, AB598 results in full inhibition of intratumoral CD39 enzymatic activity, an increase in intratumoral ATP, a decrease of extracellular CD39 on tumor cells, and ultimately, control of tumor growth. In cynomolgus monkeys, systemic dosing of AB598 results in effective enzymatic inhibition in tissues, full peripheral and tissue target engagement, and a reduction in cell surface CD39 both in tissues and in the periphery. Taken together, these data support a promising therapeutic strategy of harnessing the eATP generated by standard-of-care chemotherapies to prime the tumor microenvironment for a productive antitumor immune response.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11443198/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141155622","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
FZ-AD005, a Novel DLL3-Targeted Antibody-Drug Conjugate with Topoisomerase I Inhibitor, Shows Potent Antitumor Activity in Preclinical Models. FZ-AD005 是一种新型 DLL3 靶向抗体-药物共轭物,具有拓扑异构酶 I 抑制剂,在临床前模型中显示出强大的抗肿瘤活性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.1158/1535-7163.MCT-23-0701
Qingsong Guo, Bei Gao, Ruiwen Song, Weinan Li, Shulei Zhu, Qian Xie, Sensen Lou, Lei Wang, Jiafei Shen, Teng Zhao, Yifan Zhang, Jinsong Wu, Wei Lu, Tong Yang

Delta-like ligand 3 (DLL3) is overexpressed in small cell lung cancer (SCLC) and has been considered an attractive target for SCLC therapy. Rovalpituzumab tesirine was the first DLL3-targeted antibody-drug conjugate (ADC) to enter clinical studies. However, serious adverse events limited progress in the treatment of SCLC with rovalpituzumab tesirine. In this study, we developed a novel DLL3-targeted ADC, FZ-AD005, by using DXd with potent cytotoxicity and a relatively better safety profile to maximize the therapeutic index. FZ-AD005 was generated by a novel anti-DLL3 antibody, FZ-A038, and a valine-alanine (Val-Ala) dipeptide linker to conjugate DXd. Moreover, Fc-silencing technology was introduced in FZ-AD005 to avoid off-target toxicity mediated by FcγRs and showed negligible Fc-mediated effector functions in vitro. In preclinical evaluation, FZ-AD005 exhibited DLL3-specific binding and demonstrated efficient internalization, bystander killing, and excellent in vivo antitumor activities in cell line-derived xenograft and patient-derived xenograft models. FZ-AD005 was stable in circulation with acceptable pharmacokinetic profiles in cynomolgus monkeys. FZ-AD005 was well tolerated in rats and monkeys. The safety profile of FZ-AD005 was favorable, and the highest nonseverely toxic dose was 30 mg/kg in cynomolgus monkeys. In conclusion, FZ-AD005 has the potential to be a superior DLL3-targeted ADC with a wide therapeutic window and is expected to provide clinical benefits for the treatment of patients with SCLC.

Delta样配体3(DLL3)在小细胞肺癌(SCLC)中过度表达,一直被认为是治疗SCLC的诱人靶点。Rovalpituzumab tesirine(Rova-T)是第一个进入临床研究的 DLL3 靶向抗体-药物共轭物(ADC)。然而,严重的不良反应限制了Rova-T治疗SCLC的进展。在这项研究中,我们利用具有强大细胞毒性和相对较好安全性的DXd开发了一种新型DLL-3靶向ADC--FZ-AD005,以最大限度地提高治疗指数。FZ-AD005由新型抗DLL3抗体FZ-A038和缬氨酸-丙氨酸(Val-Ala)二肽连接体共轭DXd制成。此外,FZ-AD005 还引入了 Fc 沉默技术,以避免 FcγRs 介导的脱靶毒性,并在体外显示出可忽略的 Fc 介导的效应功能。在临床前评估中,FZ-AD005 表现出 DLL3 特异性结合,并在细胞系衍生异种移植(CDX)和患者衍生异种移植(PDX)模型中表现出高效的内化、旁观者杀伤和出色的体内抗肿瘤活性。FZ-AD005 在犬科猴体内循环稳定,药代动力学特征可接受。大鼠和猴子对 FZ-AD005 的耐受性良好。FZ-AD005 的安全性良好,在猴体内的最高非剧毒剂量为 30 毫克/千克。总之,FZ-AD005 有可能成为一种出色的 DLL3 靶向 ADC,具有广泛的治疗窗口期,有望为治疗 SCLC 患者带来临床益处。
{"title":"FZ-AD005, a Novel DLL3-Targeted Antibody-Drug Conjugate with Topoisomerase I Inhibitor, Shows Potent Antitumor Activity in Preclinical Models.","authors":"Qingsong Guo, Bei Gao, Ruiwen Song, Weinan Li, Shulei Zhu, Qian Xie, Sensen Lou, Lei Wang, Jiafei Shen, Teng Zhao, Yifan Zhang, Jinsong Wu, Wei Lu, Tong Yang","doi":"10.1158/1535-7163.MCT-23-0701","DOIUrl":"10.1158/1535-7163.MCT-23-0701","url":null,"abstract":"<p><p>Delta-like ligand 3 (DLL3) is overexpressed in small cell lung cancer (SCLC) and has been considered an attractive target for SCLC therapy. Rovalpituzumab tesirine was the first DLL3-targeted antibody-drug conjugate (ADC) to enter clinical studies. However, serious adverse events limited progress in the treatment of SCLC with rovalpituzumab tesirine. In this study, we developed a novel DLL3-targeted ADC, FZ-AD005, by using DXd with potent cytotoxicity and a relatively better safety profile to maximize the therapeutic index. FZ-AD005 was generated by a novel anti-DLL3 antibody, FZ-A038, and a valine-alanine (Val-Ala) dipeptide linker to conjugate DXd. Moreover, Fc-silencing technology was introduced in FZ-AD005 to avoid off-target toxicity mediated by FcγRs and showed negligible Fc-mediated effector functions in vitro. In preclinical evaluation, FZ-AD005 exhibited DLL3-specific binding and demonstrated efficient internalization, bystander killing, and excellent in vivo antitumor activities in cell line-derived xenograft and patient-derived xenograft models. FZ-AD005 was stable in circulation with acceptable pharmacokinetic profiles in cynomolgus monkeys. FZ-AD005 was well tolerated in rats and monkeys. The safety profile of FZ-AD005 was favorable, and the highest nonseverely toxic dose was 30 mg/kg in cynomolgus monkeys. In conclusion, FZ-AD005 has the potential to be a superior DLL3-targeted ADC with a wide therapeutic window and is expected to provide clinical benefits for the treatment of patients with SCLC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11443207/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PET Imaging Using 89Zr Labeled StarPEG Nanocarriers Reveals Heterogeneous Enhanced Permeability and Retention (EPR) in Prostate Cancer. 使用 89Zr 标记的 StarPEG 纳米载体进行 PET 成像显示前列腺癌的异质性增强渗透性和滞留性 (EPR)。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-27 DOI: 10.1158/1535-7163.MCT-24-0024
Niranjan Meher, Anil P Bidkar, Anju Wadhwa, Kondapa Naidu Bobba, Suchi Dhrona, Chandrashekhar Dasari, Changhua Mu, Cyril O Y Fong, Juan A Cámara, Umama Ali, Megha Basak, David Bulkley, Veronica Steri, Shaun D Fontaine, Jun Zhu, Adam Oskowitz, Rahul R Aggarwal, Renuka Sriram, Jonathan Chou, David M Wilson, Youngho Seo, Daniel V Santi, Gary W Ashley, Henry F VanBrocklin, Robert R Flavell

The enhanced permeability and retention (EPR) effect controls passive nanodrug uptake in tumors, and may provide a high tumor payload with prolonged retention for cancer treatment. However, EPR-mediated tumor uptake and distribution vary by cancer phenotype. Thus, we hypothesized that a companion PET-imaging surrogate may benefit EPR-mediated therapeutic drug delivery. We developed two 89Zr-radiolabeled nanocarriers based on 4-armed-starPEG40kDa with or without talazoparib (TLZ), a potent PARPi, as surrogates for the PEG-TLZ4 therapeutic scaffold. For PET imaging, PEG-DFB4 and PEG-DFB1-TLZ3 were radiolabeled with 89Zr by replacing one or all four TLZ on PEG-TLZ4 with deferoxamine B (DFB). The radiolabeled nanodrugs [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 were tested in vivo in prostate cancer subcutaneous xenografts (22Rv1, LTL-545, and LTL-610) and 22Rv1 metastatic models. Their EPR-mediated tumoral uptake and penetration was compared to CT26, a known EPR-high MicroPET/CT images, organ biodistribution, and calculated kinetic parameters showed high uptake in CT26 and LTL-545, moderate to low uptake in LTL-610 and 22Rv1. MicroPET/CT and high-resolution autoradiographic images showed nanocarrier penetration into highly permeable CT26, but heterogeneous peripheral accumulation was observed in LTL-545, LTL-610, and 22Rv1 subcutaneous xenografts and metastatic tumors. CD31 staining of tumor sections showed homogenous vascular development in CT26 tumors and heterogeneity in other xenografts. Both [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 showed similar accumulation and distribution in subcutaneous and metastatic tumor models. Both nanocarriers can measure tumor model passive uptake heterogeneity. Although heterogeneous, prostate cancer xenografts had low EPR. These starPEG nanocarriers could be used as PET imaging surrogates to predict drug delivery and efficacy.

增强的渗透性和滞留(EPR)效应控制着肿瘤对纳米药物的被动摄取,可为癌症治疗提供较高的肿瘤有效载荷和较长的滞留时间。然而,EPR 介导的肿瘤摄取和分布因癌症表型而异。因此,我们假设配套的 PET 成像替代物可能有利于 EPR 介导的治疗药物递送。我们开发了两种基于 4-armed-starPEG40kDa 的 89Zr 放射标记纳米载体,作为 PEG-TLZ4 治疗支架的替代物,其中包含或不包含一种强效 PARPi--talazoparib(TLZ)。为了进行 PET 成像,用去氧胺 B (DFB) 替代了 PEG-TLZ4 上的一个或全部四个 TLZ,用 89Zr 对 PEG-DFB4 和 PEG-DFB1-TLZ3 进行了放射性标记。放射性标记的纳米药物 [89Zr]PEG-DFB4 和 [89Zr]PEG-DFB1-TLZ3 在前列腺癌皮下异种移植(22Rv1、LTL-545 和 LTL-610)和 22Rv1 转移模型中进行了体内试验。它们的 EPR 介导的肿瘤摄取和穿透与 CT26 进行了比较,CT26 是一种已知的 EPR 高的药物。MicroPET/CT 图像、器官生物分布和计算的动力学参数显示 CT26 和 LTL-545 的摄取率高,LTL-610 和 22Rv1 的摄取率中等至低。MicroPET/CT 和高分辨率自显影图像显示纳米载体渗透到高渗透性 CT26 中,但在 LTL-545、LTL-610 和 22Rv1 皮下异种移植物和转移性肿瘤中观察到异质性外周聚集。肿瘤切片的 CD31 染色显示,CT26 肿瘤中的血管发育是同质的,而其他异种移植物中的血管发育则是异质的。[89Zr]PEG-DFB4和[89Zr]PEG-DFB1-TLZ3在皮下和转移性肿瘤模型中都显示出相似的聚集和分布。这两种纳米载体都能测量肿瘤模型被动摄取的异质性。尽管存在异质性,但前列腺癌异种移植物的 EPR 较低。这些星形 PEG 纳米载体可用作 PET 成像替代物来预测药物的输送和疗效。
{"title":"PET Imaging Using 89Zr Labeled StarPEG Nanocarriers Reveals Heterogeneous Enhanced Permeability and Retention (EPR) in Prostate Cancer.","authors":"Niranjan Meher, Anil P Bidkar, Anju Wadhwa, Kondapa Naidu Bobba, Suchi Dhrona, Chandrashekhar Dasari, Changhua Mu, Cyril O Y Fong, Juan A Cámara, Umama Ali, Megha Basak, David Bulkley, Veronica Steri, Shaun D Fontaine, Jun Zhu, Adam Oskowitz, Rahul R Aggarwal, Renuka Sriram, Jonathan Chou, David M Wilson, Youngho Seo, Daniel V Santi, Gary W Ashley, Henry F VanBrocklin, Robert R Flavell","doi":"10.1158/1535-7163.MCT-24-0024","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0024","url":null,"abstract":"<p><p>The enhanced permeability and retention (EPR) effect controls passive nanodrug uptake in tumors, and may provide a high tumor payload with prolonged retention for cancer treatment. However, EPR-mediated tumor uptake and distribution vary by cancer phenotype. Thus, we hypothesized that a companion PET-imaging surrogate may benefit EPR-mediated therapeutic drug delivery. We developed two 89Zr-radiolabeled nanocarriers based on 4-armed-starPEG40kDa with or without talazoparib (TLZ), a potent PARPi, as surrogates for the PEG-TLZ4 therapeutic scaffold. For PET imaging, PEG-DFB4 and PEG-DFB1-TLZ3 were radiolabeled with 89Zr by replacing one or all four TLZ on PEG-TLZ4 with deferoxamine B (DFB). The radiolabeled nanodrugs [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 were tested in vivo in prostate cancer subcutaneous xenografts (22Rv1, LTL-545, and LTL-610) and 22Rv1 metastatic models. Their EPR-mediated tumoral uptake and penetration was compared to CT26, a known EPR-high MicroPET/CT images, organ biodistribution, and calculated kinetic parameters showed high uptake in CT26 and LTL-545, moderate to low uptake in LTL-610 and 22Rv1. MicroPET/CT and high-resolution autoradiographic images showed nanocarrier penetration into highly permeable CT26, but heterogeneous peripheral accumulation was observed in LTL-545, LTL-610, and 22Rv1 subcutaneous xenografts and metastatic tumors. CD31 staining of tumor sections showed homogenous vascular development in CT26 tumors and heterogeneity in other xenografts. Both [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 showed similar accumulation and distribution in subcutaneous and metastatic tumor models. Both nanocarriers can measure tumor model passive uptake heterogeneity. Although heterogeneous, prostate cancer xenografts had low EPR. These starPEG nanocarriers could be used as PET imaging surrogates to predict drug delivery and efficacy.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142350348","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PARP7 Inhibitors and AHR Agonists Act Synergistically Across a Wide-Range of Cancer Models. PARP7 抑制剂和 AHR 激动剂在多种癌症模型中发挥协同作用
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-24 DOI: 10.1158/1535-7163.MCT-24-0211
Huadong Chen, Xuxu Gou, Ying Mao, Patrick C O'Leary, Morgan E Diolaiti, Alan Ashworth

Small molecule inhibitors of the mono (ADP) ribosyl transferase PARP7 are being evaluated as a monotherapy for tumors overexpressing PARP7 and in combination with immune checkpoint blockade. We previously showed that sensitivity to the PARP7 inhibitor (PARP7i) RBN-2397 could be enhanced by co-treatment with agonists of the Aryl Hydrocarbon Receptor (AHRa) in cell lines that show strong intrinsic sensitivity to RBN-2397. Here we demonstrate that a range of tumor cell lines that are relatively insensitive to PARP7i or AHRa as individual agents are unexpectedly profoundly sensitive to the combination. Our data show that this synergistic response is dependent on AHR/ARNT and is associated with increased levels of nuclear AHR and increased transcription of AHR target genes. In some hormone receptor-positive cell lines, we find that combination treatment is associated with proteasomal turnover of the steroid hormone receptors, androgen receptor and estrogen receptor. Both wildtype and hormone-resistant mutant forms of these receptors are degraded upon treatment with AHRa and PARP7i in breast and prostate cancer models. These results suggest that combining PARP7i with AHRa may extend the utility of these drugs to a wider range of tumors, including those that are refractory to hormone therapy.

目前正在评估单(ADP)核糖转移酶PARP7的小分子抑制剂,将其作为治疗过表达PARP7肿瘤的单一疗法,或与免疫检查点阻断疗法联合使用。我们以前的研究表明,在对 RBN-2397 表现出强烈内在敏感性的细胞系中,通过与芳基烃受体(AHRa)激动剂联合处理,可以提高 PARP7 抑制剂(PARP7i)RBN-2397 的敏感性。在这里,我们证明了一系列对 PARP7i 或 AHRa 单药相对不敏感的肿瘤细胞系意外地对联合用药非常敏感。我们的数据显示,这种协同反应依赖于 AHR/ARNT,并与核 AHR 水平的增加和 AHR 靶基因转录的增加有关。在一些激素受体阳性细胞系中,我们发现联合治疗与类固醇激素受体、雄激素受体和雌激素受体的蛋白酶体转换有关。在乳腺癌和前列腺癌模型中,使用 AHRa 和 PARP7i 治疗后,这些受体的野生型和激素抗性突变型都会降解。这些结果表明,将 PARP7i 与 AHRa 结合使用可将这些药物的用途扩大到更广泛的肿瘤,包括那些对激素疗法难治的肿瘤。
{"title":"PARP7 Inhibitors and AHR Agonists Act Synergistically Across a Wide-Range of Cancer Models.","authors":"Huadong Chen, Xuxu Gou, Ying Mao, Patrick C O'Leary, Morgan E Diolaiti, Alan Ashworth","doi":"10.1158/1535-7163.MCT-24-0211","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0211","url":null,"abstract":"<p><p>Small molecule inhibitors of the mono (ADP) ribosyl transferase PARP7 are being evaluated as a monotherapy for tumors overexpressing PARP7 and in combination with immune checkpoint blockade. We previously showed that sensitivity to the PARP7 inhibitor (PARP7i) RBN-2397 could be enhanced by co-treatment with agonists of the Aryl Hydrocarbon Receptor (AHRa) in cell lines that show strong intrinsic sensitivity to RBN-2397. Here we demonstrate that a range of tumor cell lines that are relatively insensitive to PARP7i or AHRa as individual agents are unexpectedly profoundly sensitive to the combination. Our data show that this synergistic response is dependent on AHR/ARNT and is associated with increased levels of nuclear AHR and increased transcription of AHR target genes. In some hormone receptor-positive cell lines, we find that combination treatment is associated with proteasomal turnover of the steroid hormone receptors, androgen receptor and estrogen receptor. Both wildtype and hormone-resistant mutant forms of these receptors are degraded upon treatment with AHRa and PARP7i in breast and prostate cancer models. These results suggest that combining PARP7i with AHRa may extend the utility of these drugs to a wider range of tumors, including those that are refractory to hormone therapy.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142308092","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrated imaging probe and bispecific antibody development enables in vivo targeting of glypican-3-expressing hepatocellular carcinoma. 集成成像探针和双特异性抗体开发,实现体内靶向治疗表达 glypican-3 的肝细胞癌。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-23 DOI: 10.1158/1535-7163.MCT-23-0470
Peiman Habibollahi, Alexey Gurevich, James Z Hui, Kelley Weinfurtner, George McClung, Justin Adler, Michael C Soulen, David E Kaplan, Gregory J Nadolski, Stephen J Hunt, Andrew Tsourkas, Terence P Gade

Glypican-3 (GPC3) is a proteoglycan with high sensitivity and specificity for hepatocellular carcinoma (HCC). We describe the integrated development and validation of a GPC3-targeting optical imaging probe and T-cell redirecting antibody (TRAB) as a theranostic strategy for the detection and treatment of HCC. A novel TRAB targeting GPC3 on HCC tumor cells and the CD3 T-cell receptor as well as a distinct GPC3-specific optical imaging probe were developed from a short peptide. The efficacy of GPC3/CD3 TRAB was evaluated in vitro using interferon-γ release and calcein-AM assays. Patient-derived xenografts (PDX) were used to assess the in vivo efficacy of GPC3/CD3 TRAB and the GPC3 imaging probe for the detection of GPC3+ HCC. GPC3/CD3 TRAB caused a dose-dependent escalation in interferon-γ release from inactive peripheral blood T-cells (P = 0.001) and higher tumor-cell lysis (P = 0.01) compared to controls in vitro. Intratumorally injected GPC3/CD3 TRAB resulted in significant prolongation of tumor doubling time in the GPC3+ PDX mice, with an associated reduction of tumor fluorescent signal from the HiLyte 488- conjugated GPC3 specific peptide on optical imaging. HCC cell targeting using a GPC3/CD3 TRAB derived from a small peptide resulted in effective T-cell activation and induction of a cytotoxic response toward GPC3+ HCC tumor cells both in vitro and in vivo. GPC3-specific optical imaging enabled the detection of the GPC3+ HCC cells and noninvasive monitoring of tumor response to adoptive immunotherapy. The integrated development of a targeted therapeutic and molecular imaging probe provides a novel paradigm for developing cancer theranostics.

Glypican-3(GPC3)是一种蛋白聚糖,对肝细胞癌(HCC)具有高灵敏度和特异性。我们介绍了 GPC3 靶向光学成像探针和 T 细胞重定向抗体(TRAB)作为检测和治疗 HCC 的治疗策略的综合开发和验证。研究人员利用一种短肽开发出了一种新型的针对 HCC 肿瘤细胞上的 GPC3 和 CD3 T 细胞受体的 TRAB 以及一种独特的 GPC3 特异性光学成像探针。利用干扰素-γ释放和钙黄绿素-AM测定法在体外评估了GPC3/CD3 TRAB的疗效。患者衍生异种移植(PDX)用于评估 GPC3/CD3 TRAB 和 GPC3 成像探针在检测 GPC3+ HCC 方面的体内疗效。与体外对照组相比,GPC3/CD3 TRAB 可使非活性外周血 T 细胞释放的干扰素-γ 呈剂量依赖性增加(P = 0.001),肿瘤细胞裂解率更高(P = 0.01)。瘤内注射 GPC3/CD3 TRAB 可显著延长 GPC3+ PDX 小鼠的肿瘤倍增时间,同时在光学成像中,HiLyte 488 共轭 GPC3 特异性肽的肿瘤荧光信号也会随之减少。使用源自小肽的 GPC3/CD3 TRAB 靶向 HCC 细胞可有效激活 T 细胞,并在体外和体内诱导针对 GPC3+ HCC 肿瘤细胞的细胞毒反应。GPC3特异性光学成像能够检测GPC3+ HCC细胞,并对肿瘤对采纳性免疫疗法的反应进行无创监测。靶向治疗和分子成像探针的综合开发为开发癌症治疗技术提供了一种新的范例。
{"title":"Integrated imaging probe and bispecific antibody development enables in vivo targeting of glypican-3-expressing hepatocellular carcinoma.","authors":"Peiman Habibollahi, Alexey Gurevich, James Z Hui, Kelley Weinfurtner, George McClung, Justin Adler, Michael C Soulen, David E Kaplan, Gregory J Nadolski, Stephen J Hunt, Andrew Tsourkas, Terence P Gade","doi":"10.1158/1535-7163.MCT-23-0470","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-23-0470","url":null,"abstract":"<p><p>Glypican-3 (GPC3) is a proteoglycan with high sensitivity and specificity for hepatocellular carcinoma (HCC). We describe the integrated development and validation of a GPC3-targeting optical imaging probe and T-cell redirecting antibody (TRAB) as a theranostic strategy for the detection and treatment of HCC. A novel TRAB targeting GPC3 on HCC tumor cells and the CD3 T-cell receptor as well as a distinct GPC3-specific optical imaging probe were developed from a short peptide. The efficacy of GPC3/CD3 TRAB was evaluated in vitro using interferon-γ release and calcein-AM assays. Patient-derived xenografts (PDX) were used to assess the in vivo efficacy of GPC3/CD3 TRAB and the GPC3 imaging probe for the detection of GPC3+ HCC. GPC3/CD3 TRAB caused a dose-dependent escalation in interferon-γ release from inactive peripheral blood T-cells (P = 0.001) and higher tumor-cell lysis (P = 0.01) compared to controls in vitro. Intratumorally injected GPC3/CD3 TRAB resulted in significant prolongation of tumor doubling time in the GPC3+ PDX mice, with an associated reduction of tumor fluorescent signal from the HiLyte 488- conjugated GPC3 specific peptide on optical imaging. HCC cell targeting using a GPC3/CD3 TRAB derived from a small peptide resulted in effective T-cell activation and induction of a cytotoxic response toward GPC3+ HCC tumor cells both in vitro and in vivo. GPC3-specific optical imaging enabled the detection of the GPC3+ HCC cells and noninvasive monitoring of tumor response to adoptive immunotherapy. The integrated development of a targeted therapeutic and molecular imaging probe provides a novel paradigm for developing cancer theranostics.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291434","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Looking Beyond Checkpoint Inhibitor Monotherapy: Uncovering New Frontiers for Pancreatic Cancer Immunotherapy. 超越检查点抑制剂单药治疗:探索胰腺癌免疫疗法的新前沿。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-23 DOI: 10.1158/1535-7163.MCT-24-0311
Kajal Gupta, Eileena F Giurini, Oliver Ralph, Sam G Pappas

Pancreatic ductal adenocarcinoma (PDAC) stands out as one of the most aggressive and challenging tumors, characterized by a bleak prognosis with a mere 11% survival rate over five years in the United States. Its formidable nature is primarily attributed to its highly aggressive behavior and poor response to existing therapies. PDAC, being notably resistant to immune interventions, presents a significant obstacle in treatment strategies. While immune checkpoint inhibitor therapies have revolutionized outcomes for various cancers, their efficacy in PDAC remains exceedingly low, benefiting less than 1% of patients. The consistent failure of these therapies in PDAC has prompted intensive investigation, particularly at the preclinical level, to unravel the intricate mechanisms of resistance inherent in this cancer type. This pursuit aims to pave the way for the development of novel immunotherapeutic strategies tailored to the distinct characteristics of PDAC. This review endeavors to provide a comprehensive exploration of these emerging immunotherapy approaches in PDAC, with a specific emphasis on elucidating their underlying immunological mechanisms. Additionally, it sheds light on recently identified factors driving resistance to immunotherapy and evasion of the immune system in PDAC, offering insights beyond the conventional drivers that have been extensively studied.

胰腺导管腺癌(PDAC)是侵袭性最强、最具挑战性的肿瘤之一,其特点是预后凄惨,在美国五年以上的存活率仅为 11%。它的可怕之处主要在于其高度侵袭性和对现有疗法的不良反应。PDAC 对免疫干预具有明显的抗药性,给治疗策略带来了巨大障碍。虽然免疫检查点抑制剂疗法彻底改变了各种癌症的治疗效果,但其对 PDAC 的疗效仍然非常低,只有不到 1%的患者从中受益。这些疗法在 PDAC 中的持续失败促使人们进行深入研究,特别是在临床前水平,以揭示这种癌症类型固有的复杂耐药机制。这一研究旨在为开发针对 PDAC 独特特征的新型免疫治疗策略铺平道路。本综述旨在全面探讨这些针对 PDAC 的新兴免疫疗法,特别强调阐明其潜在的免疫机制。此外,它还揭示了新近发现的 PDAC 免疫疗法耐药和免疫系统逃避的因素,提供了超越已被广泛研究的传统驱动因素的见解。
{"title":"Looking Beyond Checkpoint Inhibitor Monotherapy: Uncovering New Frontiers for Pancreatic Cancer Immunotherapy.","authors":"Kajal Gupta, Eileena F Giurini, Oliver Ralph, Sam G Pappas","doi":"10.1158/1535-7163.MCT-24-0311","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0311","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) stands out as one of the most aggressive and challenging tumors, characterized by a bleak prognosis with a mere 11% survival rate over five years in the United States. Its formidable nature is primarily attributed to its highly aggressive behavior and poor response to existing therapies. PDAC, being notably resistant to immune interventions, presents a significant obstacle in treatment strategies. While immune checkpoint inhibitor therapies have revolutionized outcomes for various cancers, their efficacy in PDAC remains exceedingly low, benefiting less than 1% of patients. The consistent failure of these therapies in PDAC has prompted intensive investigation, particularly at the preclinical level, to unravel the intricate mechanisms of resistance inherent in this cancer type. This pursuit aims to pave the way for the development of novel immunotherapeutic strategies tailored to the distinct characteristics of PDAC. This review endeavors to provide a comprehensive exploration of these emerging immunotherapy approaches in PDAC, with a specific emphasis on elucidating their underlying immunological mechanisms. Additionally, it sheds light on recently identified factors driving resistance to immunotherapy and evasion of the immune system in PDAC, offering insights beyond the conventional drivers that have been extensively studied.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291497","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A B7-H3-targeted CD28 bispecific antibody enhances the activity of anti-PD1 and CD3 T-cell engager immunotherapies. B7-H3 靶向 CD28 双特异性抗体可增强抗 PD1 和 CD3 T 细胞吸引免疫疗法的活性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-20 DOI: 10.1158/1535-7163.MCT-24-0327
Gregory L Moore, Veronica G Zeng, Juan E Diaz, Christine Bonzon, Kendra N Avery, Rumana Rashid, Jing Qi, Dong Hyun Nam, Jonathan Jacinto, Matthew A Dragovich, Yoon Kyung Kim, Karen P Balcazar, Charles G Bakhit, Araz Eivazi, Hanh Nguyen, Umesh S Muchhal, David E Szymkowski, John R Desjarlais, Michael Hedvat

T-cell activation is a multistep process requiring T-cell receptor engagement by peptide-major histocompatibility complexes (Signal 1) coupled with CD28-mediated costimulation (Signal 2). Tumors typically lack expression of CD28 ligands, so tumor-specific Signal 1 (e.g., neoepitope presentation) without costimulation may be ineffective or even induce T-cell anergy. We designed the bispecific antibody XmAb808 to co-engage the tumor-associated antigen B7-H3 with CD28 to promote T-cell costimulation within the tumor microenvironment. XmAb808 costimulation was measured by its ability to activate and expand T cells and enhance T cell-mediated cancer cell killing in cocultures of human peripheral blood mononuclear cells (PBMCs) and cancer cells, and in mice engrafted with human PBMCs and tumor xenografts. XmAb808 avidly bound cancer cells and stimulated interleukin (IL)2 and interferon (IFN)γ secretion from T cells cocultured with cancer cells engineered to deliver Signal 1 to T cells via a surface-expressed anti-CD3 antibody. XmAb808 enhanced expression of the anti-apoptotic factor Bcl-xL and CD25, promoting survival and IL2-dependent expansion of T cells coupled with increased T cell-mediated cytotoxicity in vitro. XmAb808 combined with a EpCAM×CD3 bispecific antibody to enhance target cell killing through IL2-dependent expansion of CD25+ T cells. This combination also suppressed pancreatic tumor xenograft growth in mice. Further, XmAb808 combined with an anti-PD1 antibody to suppress breast tumor xenograft growth in mice. XmAb808 as monotherapy and in combination with an anti-PD1 antibody is currently in clinical development in patients with advanced solid tumors. Our results suggest that XmAb808 may also combine with tumor antigen-targeted anti-CD3 (Signal 1) T-cell engagers.

T 细胞活化是一个多步骤过程,需要肽-主要组织相容性复合物(信号 1)与 CD28 介导的成本刺激(信号 2)作用于 T 细胞受体。肿瘤通常缺乏 CD28 配体的表达,因此没有成本刺激的肿瘤特异性信号 1(如新表位呈现)可能无效,甚至会诱发 T 细胞过敏。我们设计了双特异性抗体 XmAb808,使肿瘤相关抗原 B7-H3 与 CD28 共同结合,促进肿瘤微环境中的 T 细胞成本刺激。在人外周血单核细胞(PBMCs)和癌细胞的共培养物中,以及在移植了人PBMCs和肿瘤异种移植物的小鼠体内,XmAb808的成本刺激能力是通过其激活和扩增T细胞以及增强T细胞介导的癌细胞杀伤能力来衡量的。XmAb808 能亲和癌细胞,刺激与癌细胞共培养的 T 细胞分泌白细胞介素 (IL)2 和干扰素 (IFN)γ,这些癌细胞通过表面表达的抗 CD3 抗体向 T 细胞传递信号 1。XmAb808 提高了抗凋亡因子 Bcl-xL 和 CD25 的表达,促进了 T 细胞的存活和 IL2 依赖性扩增,同时提高了 T 细胞介导的体外细胞毒性。XmAb808 与 EpCAM×CD3 双特异性抗体相结合,可通过依赖 IL2 的 CD25+ T 细胞扩增增强对靶细胞的杀伤力。这种组合还能抑制小鼠胰腺肿瘤异种移植的生长。此外,XmAb808 还与抗 PD1 抗体相结合,抑制了乳腺肿瘤异种移植小鼠的生长。目前,XmAb808 作为单药或与抗 PD1 抗体联用正在晚期实体瘤患者中进行临床开发。我们的研究结果表明,XmAb808 还可以与肿瘤抗原靶向的抗 CD3(信号 1)T 细胞吸引剂联合使用。
{"title":"A B7-H3-targeted CD28 bispecific antibody enhances the activity of anti-PD1 and CD3 T-cell engager immunotherapies.","authors":"Gregory L Moore, Veronica G Zeng, Juan E Diaz, Christine Bonzon, Kendra N Avery, Rumana Rashid, Jing Qi, Dong Hyun Nam, Jonathan Jacinto, Matthew A Dragovich, Yoon Kyung Kim, Karen P Balcazar, Charles G Bakhit, Araz Eivazi, Hanh Nguyen, Umesh S Muchhal, David E Szymkowski, John R Desjarlais, Michael Hedvat","doi":"10.1158/1535-7163.MCT-24-0327","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0327","url":null,"abstract":"<p><p>T-cell activation is a multistep process requiring T-cell receptor engagement by peptide-major histocompatibility complexes (Signal 1) coupled with CD28-mediated costimulation (Signal 2). Tumors typically lack expression of CD28 ligands, so tumor-specific Signal 1 (e.g., neoepitope presentation) without costimulation may be ineffective or even induce T-cell anergy. We designed the bispecific antibody XmAb808 to co-engage the tumor-associated antigen B7-H3 with CD28 to promote T-cell costimulation within the tumor microenvironment. XmAb808 costimulation was measured by its ability to activate and expand T cells and enhance T cell-mediated cancer cell killing in cocultures of human peripheral blood mononuclear cells (PBMCs) and cancer cells, and in mice engrafted with human PBMCs and tumor xenografts. XmAb808 avidly bound cancer cells and stimulated interleukin (IL)2 and interferon (IFN)γ secretion from T cells cocultured with cancer cells engineered to deliver Signal 1 to T cells via a surface-expressed anti-CD3 antibody. XmAb808 enhanced expression of the anti-apoptotic factor Bcl-xL and CD25, promoting survival and IL2-dependent expansion of T cells coupled with increased T cell-mediated cytotoxicity in vitro. XmAb808 combined with a EpCAM×CD3 bispecific antibody to enhance target cell killing through IL2-dependent expansion of CD25+ T cells. This combination also suppressed pancreatic tumor xenograft growth in mice. Further, XmAb808 combined with an anti-PD1 antibody to suppress breast tumor xenograft growth in mice. XmAb808 as monotherapy and in combination with an anti-PD1 antibody is currently in clinical development in patients with advanced solid tumors. Our results suggest that XmAb808 may also combine with tumor antigen-targeted anti-CD3 (Signal 1) T-cell engagers.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291431","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advanced Human Papillomavirus-Negative Head and Neck Squamous Cell Carcinoma: Unmet Need and Emerging Therapies. 晚期人类乳头瘤病毒阴性头颈部鳞状细胞癌:未满足的需求和新兴疗法。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-09-20 DOI: 10.1158/1535-7163.MCT-24-0281
Robin Park, Christine H Chung

Despite notable progress in the treatment of advanced head and neck squamous cell carcinoma (HNSCC), survival remains poor in patients with recurrent and/or metastatic (R/M) human papillomavirus (HPV)-negative HNSCC. Worse outcomes in the HPV-negative patients may be partly related to loss of cell-cycle regulators and tumor suppressors as well as a noninflamed and hypoxic tumor microenvironment, both of which contribute to treatment resistance and disease progression. Anti-programmed cell death protein 1-based regimens as current standard-of-care treatment for R/M HNSCC are associated with durable responses in a limited number of patients. The anti-epidermal growth factor receptor (EGFR) monoclonal antibody, cetuximab, has antitumor activity in this treatment setting, but responses are short-lived and inevitably curtailed due to treatment resistance. Crosstalk between the EGFR and hepatocyte growth factor (HGF)-dependent mesenchymal-epithelial transition (c-MET) receptor tyrosine kinase pathway is a known mechanism of resistance to cetuximab. Dual targeting of EGFR and c-MET pathways may overcome resistance to cetuximab in patients with HPV-negative HNSCC. Here, we review clinical data of treatments evaluated in patients with R/M HPV-negative HNSCC and highlight the potential role of combining HGF/c-MET and EGFR pathway inhibitors to overcome cetuximab resistance in this population.

尽管晚期头颈部鳞状细胞癌(HNSCC)的治疗取得了显著进展,但复发性和/或转移性(R/M)人乳头瘤病毒(HPV)阴性 HNSCC 患者的生存率仍然很低。HPV阴性患者预后较差的部分原因可能与细胞周期调节因子和肿瘤抑制因子的缺失以及非炎症和缺氧的肿瘤微环境有关,这两种因素都会导致耐药性和疾病进展。以抗程序性细胞死亡蛋白 1 为基础的治疗方案是目前治疗 R/M HNSCC 的标准疗法,但只有少数患者能获得持久的疗效。抗表皮生长因子受体(EGFR)单克隆抗体西妥昔单抗(cetuximab)在这种治疗环境中具有抗肿瘤活性,但疗效短暂,而且不可避免地会因耐药性而减弱。表皮生长因子受体(EGFR)与肝细胞生长因子(HGF)依赖性间充质-上皮转化(c-MET)受体酪氨酸激酶通路之间的串扰是已知的西妥昔单抗耐药机制。对表皮生长因子受体和c-MET通路进行双重靶向治疗可能会克服HPV阴性HNSCC患者对西妥昔单抗的耐药性。在此,我们回顾了对R/M型HPV阴性HNSCC患者进行治疗评估的临床数据,并强调了联合HGF/c-MET和表皮生长因子受体途径抑制剂在克服该人群西妥昔单抗耐药方面的潜在作用。
{"title":"Advanced Human Papillomavirus-Negative Head and Neck Squamous Cell Carcinoma: Unmet Need and Emerging Therapies.","authors":"Robin Park, Christine H Chung","doi":"10.1158/1535-7163.MCT-24-0281","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0281","url":null,"abstract":"<p><p>Despite notable progress in the treatment of advanced head and neck squamous cell carcinoma (HNSCC), survival remains poor in patients with recurrent and/or metastatic (R/M) human papillomavirus (HPV)-negative HNSCC. Worse outcomes in the HPV-negative patients may be partly related to loss of cell-cycle regulators and tumor suppressors as well as a noninflamed and hypoxic tumor microenvironment, both of which contribute to treatment resistance and disease progression. Anti-programmed cell death protein 1-based regimens as current standard-of-care treatment for R/M HNSCC are associated with durable responses in a limited number of patients. The anti-epidermal growth factor receptor (EGFR) monoclonal antibody, cetuximab, has antitumor activity in this treatment setting, but responses are short-lived and inevitably curtailed due to treatment resistance. Crosstalk between the EGFR and hepatocyte growth factor (HGF)-dependent mesenchymal-epithelial transition (c-MET) receptor tyrosine kinase pathway is a known mechanism of resistance to cetuximab. Dual targeting of EGFR and c-MET pathways may overcome resistance to cetuximab in patients with HPV-negative HNSCC. Here, we review clinical data of treatments evaluated in patients with R/M HPV-negative HNSCC and highlight the potential role of combining HGF/c-MET and EGFR pathway inhibitors to overcome cetuximab resistance in this population.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142291432","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1