Pub Date : 2024-07-22DOI: 10.1158/2159-8290.CD-23-1323
Norihiro Yamaguchi, Y Gloria Wu, Ethan Ravetch, Mai Takahashi, Abdul G Khan, Akimasa Hayashi, Wenbin Mei, Dennis Hsu, Shigeaki Umeda, Elisa de Stanchina, Ivo C Lorenz, Christine A Iacobuzio-Donahue, Sohail F Tavazoie
Pancreatic ductal adenocarcinoma (PDAC) is an increasingly diagnosed cancer that kills 90% of afflicted patients, with most patients receiving palliative chemotherapy. We identified neuronal pentraxin 1 (NPTX1) as a cancer secreted protein that becomes over-expressed in human and murine PDAC cells during metastatic progression and identified adhesion molecule with Ig like domain 2 (AMIGO2) as its receptor. Molecular, genetic, biochemical and pharmacologic experiments revealed that secreted NPTX1 acts cell-autonomously on the AMIGO2 receptor to drive PDAC metastatic colonization of the liver-the primary site of PDAC metastasis. NPTX1-AMIGO2 signaling enhanced hypoxic growth and was critically required for hypoxia induced factor-1a (HIF1a) nuclear retention and function. NPTX1 is over-expressed in human PDAC tumors and upregulated in liver metastases. Therapeutic targeting of NPTX1 with a high-affinity monoclonal antibody substantially reduced PDAC liver metastatic colonization. We thus identify NPTX1-AMIGO2 as druggable critical upstream regulators of the HIF1a hypoxic response in PDAC.
{"title":"A targetable secreted neural protein drives pancreatic cancer metastatic colonization and HIF1a nuclear retention.","authors":"Norihiro Yamaguchi, Y Gloria Wu, Ethan Ravetch, Mai Takahashi, Abdul G Khan, Akimasa Hayashi, Wenbin Mei, Dennis Hsu, Shigeaki Umeda, Elisa de Stanchina, Ivo C Lorenz, Christine A Iacobuzio-Donahue, Sohail F Tavazoie","doi":"10.1158/2159-8290.CD-23-1323","DOIUrl":"10.1158/2159-8290.CD-23-1323","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is an increasingly diagnosed cancer that kills 90% of afflicted patients, with most patients receiving palliative chemotherapy. We identified neuronal pentraxin 1 (NPTX1) as a cancer secreted protein that becomes over-expressed in human and murine PDAC cells during metastatic progression and identified adhesion molecule with Ig like domain 2 (AMIGO2) as its receptor. Molecular, genetic, biochemical and pharmacologic experiments revealed that secreted NPTX1 acts cell-autonomously on the AMIGO2 receptor to drive PDAC metastatic colonization of the liver-the primary site of PDAC metastasis. NPTX1-AMIGO2 signaling enhanced hypoxic growth and was critically required for hypoxia induced factor-1a (HIF1a) nuclear retention and function. NPTX1 is over-expressed in human PDAC tumors and upregulated in liver metastases. Therapeutic targeting of NPTX1 with a high-affinity monoclonal antibody substantially reduced PDAC liver metastatic colonization. We thus identify NPTX1-AMIGO2 as druggable critical upstream regulators of the HIF1a hypoxic response in PDAC.</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":" ","pages":""},"PeriodicalIF":29.7,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141726945","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-24-0558
Kayla R Kulhanek, Anusha Kalbasi
Despite its long history of toxicity and limited efficacy, IL2 has re-entered the clinic as a companion to the recently FDA-approved tumor infiltrating lymphocyte therapy. In back-to-back articles, Moynihan and Kaptein introduce a new fusion protein that delivers a biased IL2 mutein to CD8 T cells. See related article by Moynihan et al., p. 1206 (6). See related article by Kaptein et al., p. 1226 (7).
{"title":"Targeted Bias: The Next Swing at IL2 Therapy.","authors":"Kayla R Kulhanek, Anusha Kalbasi","doi":"10.1158/2159-8290.CD-24-0558","DOIUrl":"10.1158/2159-8290.CD-24-0558","url":null,"abstract":"<p><p>Despite its long history of toxicity and limited efficacy, IL2 has re-entered the clinic as a companion to the recently FDA-approved tumor infiltrating lymphocyte therapy. In back-to-back articles, Moynihan and Kaptein introduce a new fusion protein that delivers a biased IL2 mutein to CD8 T cells. See related article by Moynihan et al., p. 1206 (6). See related article by Kaptein et al., p. 1226 (7).</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":"14 7","pages":"1145-1146"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466378","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-24-0128
William Hill, Clare E Weeden, Charles Swanton
Environmental carcinogens increase cancer incidence via both mutagenic and non-mutagenic mechanisms. There are over 500 known or suspected carcinogens classified by the International Agency for Research on Cancer. Sequencing of both cancerous and histologically non-cancerous tissue has been instrumental in improving our understanding of how environmental carcinogens cause cancer. Understanding how and defining which environmental or lifestyle exposures drive cancer will support cancer prevention. Recent research is revisiting the mechanisms of early tumorigenesis, paving the way for an era of molecular cancer prevention. Significance: Recent data have improved our understanding of how carcinogens cause cancer, which may reveal novel opportunities for molecular cancer prevention.
{"title":"Tumor Promoters and Opportunities for Molecular Cancer Prevention.","authors":"William Hill, Clare E Weeden, Charles Swanton","doi":"10.1158/2159-8290.CD-24-0128","DOIUrl":"10.1158/2159-8290.CD-24-0128","url":null,"abstract":"<p><p>Environmental carcinogens increase cancer incidence via both mutagenic and non-mutagenic mechanisms. There are over 500 known or suspected carcinogens classified by the International Agency for Research on Cancer. Sequencing of both cancerous and histologically non-cancerous tissue has been instrumental in improving our understanding of how environmental carcinogens cause cancer. Understanding how and defining which environmental or lifestyle exposures drive cancer will support cancer prevention. Recent research is revisiting the mechanisms of early tumorigenesis, paving the way for an era of molecular cancer prevention. Significance: Recent data have improved our understanding of how carcinogens cause cancer, which may reveal novel opportunities for molecular cancer prevention.</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":" ","pages":"1154-1160"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7616438/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141316748","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-23-0216
Matheus Henrique Dias, Anoek Friskes, Siying Wang, Joao M Fernandes Neto, Frank van Gemert, Soufiane Mourragui, Chrysa Papagianni, Hendrik J Kuiken, Sara Mainardi, Daniel Alvarez-Villanueva, Cor Lieftink, Ben Morris, Anna Dekker, Emma van Dijk, Lieke H S Wilms, Marcelo S da Silva, Robin A Jansen, Antonio Mulero-Sánchez, Elke Malzer, August Vidal, Cristina Santos, Ramón Salazar, Rosangela A M Wailemann, Thompson E P Torres, Giulia De Conti, Jonne A Raaijmakers, Petur Snaebjornsson, Shengxian Yuan, Wenxin Qin, John S Kovach, Hugo A Armelin, Hein Te Riele, Alexander van Oudenaarden, Haojie Jin, Roderick L Beijersbergen, Alberto Villanueva, Rene H Medema, Rene Bernards
Cancer homeostasis depends on a balance between activated oncogenic pathways driving tumorigenesis and engagement of stress response programs that counteract the inherent toxicity of such aberrant signaling. Although inhibition of oncogenic signaling pathways has been explored extensively, there is increasing evidence that overactivation of the same pathways can also disrupt cancer homeostasis and cause lethality. We show here that inhibition of protein phosphatase 2A (PP2A) hyperactivates multiple oncogenic pathways and engages stress responses in colon cancer cells. Genetic and compound screens identify combined inhibition of PP2A and WEE1 as synergistic in multiple cancer models by collapsing DNA replication and triggering premature mitosis followed by cell death. This combination also suppressed the growth of patient-derived tumors in vivo. Remarkably, acquired resistance to this drug combination suppressed the ability of colon cancer cells to form tumors in vivo. Our data suggest that paradoxical activation of oncogenic signaling can result in tumor-suppressive resistance. Significance: A therapy consisting of deliberate hyperactivation of oncogenic signaling combined with perturbation of the stress responses that result from this is very effective in animal models of colon cancer. Resistance to this therapy is associated with loss of oncogenic signaling and reduced oncogenic capacity, indicative of tumor-suppressive drug resistance.
癌症的稳态取决于驱动肿瘤发生的致癌通路激活与应激反应程序参与之间的平衡,应激反应程序可抵消这种异常信号传导的固有毒性。尽管抑制致癌信号通路的研究已经非常广泛,但越来越多的证据表明,过度激活同样的通路也会破坏癌症稳态并导致死亡。我们在此表明,抑制蛋白磷酸酶 2A(PP2A)会过度激活多种致癌通路,并使结肠癌细胞产生应激反应。通过基因和化合物筛选发现,联合抑制 PP2A 和 WEE1 在多种癌症模型中具有协同作用,能使 DNA 复制崩溃并引发过早的有丝分裂,继而导致细胞死亡。这种组合还能抑制患者体内肿瘤的生长。值得注意的是,获得性抗药性抑制了结肠癌细胞在体内形成肿瘤的能力。我们的数据表明,致癌信号的悖论性激活可导致抑制肿瘤的抗药性。
{"title":"Paradoxical Activation of Oncogenic Signaling as a Cancer Treatment Strategy.","authors":"Matheus Henrique Dias, Anoek Friskes, Siying Wang, Joao M Fernandes Neto, Frank van Gemert, Soufiane Mourragui, Chrysa Papagianni, Hendrik J Kuiken, Sara Mainardi, Daniel Alvarez-Villanueva, Cor Lieftink, Ben Morris, Anna Dekker, Emma van Dijk, Lieke H S Wilms, Marcelo S da Silva, Robin A Jansen, Antonio Mulero-Sánchez, Elke Malzer, August Vidal, Cristina Santos, Ramón Salazar, Rosangela A M Wailemann, Thompson E P Torres, Giulia De Conti, Jonne A Raaijmakers, Petur Snaebjornsson, Shengxian Yuan, Wenxin Qin, John S Kovach, Hugo A Armelin, Hein Te Riele, Alexander van Oudenaarden, Haojie Jin, Roderick L Beijersbergen, Alberto Villanueva, Rene H Medema, Rene Bernards","doi":"10.1158/2159-8290.CD-23-0216","DOIUrl":"10.1158/2159-8290.CD-23-0216","url":null,"abstract":"<p><p>Cancer homeostasis depends on a balance between activated oncogenic pathways driving tumorigenesis and engagement of stress response programs that counteract the inherent toxicity of such aberrant signaling. Although inhibition of oncogenic signaling pathways has been explored extensively, there is increasing evidence that overactivation of the same pathways can also disrupt cancer homeostasis and cause lethality. We show here that inhibition of protein phosphatase 2A (PP2A) hyperactivates multiple oncogenic pathways and engages stress responses in colon cancer cells. Genetic and compound screens identify combined inhibition of PP2A and WEE1 as synergistic in multiple cancer models by collapsing DNA replication and triggering premature mitosis followed by cell death. This combination also suppressed the growth of patient-derived tumors in vivo. Remarkably, acquired resistance to this drug combination suppressed the ability of colon cancer cells to form tumors in vivo. Our data suggest that paradoxical activation of oncogenic signaling can result in tumor-suppressive resistance. Significance: A therapy consisting of deliberate hyperactivation of oncogenic signaling combined with perturbation of the stress responses that result from this is very effective in animal models of colon cancer. Resistance to this therapy is associated with loss of oncogenic signaling and reduced oncogenic capacity, indicative of tumor-suppressive drug resistance.</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":" ","pages":"1276-1301"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11215412/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140292881","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-23-1263
Paulien Kaptein, Nadine Slingerland, Christina Metoikidou, Felix Prinz, Simone Brokamp, Mercedes Machuca-Ostos, Guido de Roo, Ton N M Schumacher, Yik A Yeung, Kelly D Moynihan, Ivana M Djuretic, Daniela S Thommen
Tumor-specific CD8+ T cells are key effectors of antitumor immunity but are often rendered dysfunctional in the tumor microenvironment. Immune-checkpoint blockade can restore antitumor T-cell function in some patients; however, most do not respond to this therapy, often despite T-cell infiltration in their tumors. We here explored a CD8-targeted IL2 fusion molecule (CD8-IL2) to selectively reactivate intratumoral CD8+ T cells in patient-derived tumor fragments. Treatment with CD8-IL2 broadly armed intratumoral CD8+ T cells with enhanced effector capacity, thereby specifically enabling reinvigoration of the dysfunctional T-cell pool to elicit potent immune activity. Notably, the revival of dysfunctional T cells to mediate effector activity by CD8-IL2 depended on simultaneous antigen recognition and was quantitatively and qualitatively superior to that achieved by PD-1 blockade. Finally, CD8-IL2 was able to functionally reinvigorate T cells in tumors resistant to anti-PD-1, underscoring its potential as a novel treatment strategy for patients with cancer. Significance: Reinvigorating T cells is crucial for response to checkpoint blockade therapy. However, emerging evidence suggests that the PD-1/PD-L1 axis is not the sole impediment for activating T cells within tumors. Selectively targeting cytokines toward specific T-cell subsets might overcome these barriers and stimulate T cells within resistant tumors. See related article by Moynihan et al., p. 1206 (32).
肿瘤特异性 CD8+ T 细胞是抗肿瘤免疫的关键效应器,但在肿瘤微环境中往往功能失调。免疫检查点阻断疗法可以恢复部分患者的抗肿瘤T细胞功能,但大多数患者对这种疗法没有反应,尽管肿瘤中往往有T细胞浸润。我们在此研究了一种 CD8 靶向 IL2 融合分子(CD8-IL2),以选择性地重新激活患者肿瘤片段中的瘤内 CD8+ T 细胞。用CD8-IL2治疗可广泛武装瘤内CD8+ T细胞,增强其效应能力,从而特异性地使功能失调的T细胞池重获活力,激发强大的免疫活性。值得注意的是,CD8-IL2 能使功能失调的 T 细胞恢复介导效应活性,这取决于同时进行的抗原识别,而且在数量和质量上都优于 PD-1 阻断疗法。最后,CD8-IL2能够在抗PD-1的肿瘤中重新激活T细胞的功能,突出了其作为癌症患者新型治疗策略的潜力。
{"title":"CD8-Targeted IL2 Unleashes Tumor-Specific Immunity in Human Cancer Tissue by Reviving the Dysfunctional T-cell Pool.","authors":"Paulien Kaptein, Nadine Slingerland, Christina Metoikidou, Felix Prinz, Simone Brokamp, Mercedes Machuca-Ostos, Guido de Roo, Ton N M Schumacher, Yik A Yeung, Kelly D Moynihan, Ivana M Djuretic, Daniela S Thommen","doi":"10.1158/2159-8290.CD-23-1263","DOIUrl":"10.1158/2159-8290.CD-23-1263","url":null,"abstract":"<p><p>Tumor-specific CD8+ T cells are key effectors of antitumor immunity but are often rendered dysfunctional in the tumor microenvironment. Immune-checkpoint blockade can restore antitumor T-cell function in some patients; however, most do not respond to this therapy, often despite T-cell infiltration in their tumors. We here explored a CD8-targeted IL2 fusion molecule (CD8-IL2) to selectively reactivate intratumoral CD8+ T cells in patient-derived tumor fragments. Treatment with CD8-IL2 broadly armed intratumoral CD8+ T cells with enhanced effector capacity, thereby specifically enabling reinvigoration of the dysfunctional T-cell pool to elicit potent immune activity. Notably, the revival of dysfunctional T cells to mediate effector activity by CD8-IL2 depended on simultaneous antigen recognition and was quantitatively and qualitatively superior to that achieved by PD-1 blockade. Finally, CD8-IL2 was able to functionally reinvigorate T cells in tumors resistant to anti-PD-1, underscoring its potential as a novel treatment strategy for patients with cancer. Significance: Reinvigorating T cells is crucial for response to checkpoint blockade therapy. However, emerging evidence suggests that the PD-1/PD-L1 axis is not the sole impediment for activating T cells within tumors. Selectively targeting cytokines toward specific T-cell subsets might overcome these barriers and stimulate T cells within resistant tumors. See related article by Moynihan et al., p. 1206 (32).</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":" ","pages":"1226-1251"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11215409/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140334811","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-24-0377
Javier Carmona, Elena Chavarria, Kate Donoghue, Christina von Gertten, Petra Oberrauch, Emma Pailler, Giovanni Scoazec, Ruud Weijer, Judith Balmaña, Irene Brana, Cinzia Brunelli, Suzette Delaloge, Marc Deloger, Pierre Delpy, Ingemar Ernberg, Rebecca C Fitzgerald, Elena Garralda, Martin Lablans, Janne Lëhtio, Carlos Lopez, Maialen Fernández, Rosalba Miceli, Paolo Nuciforo, Raquel Perez-Lopez, Elena Provenzano, Marjanka K Schmidt, Cesar Serrano, Neeltje Steeghs, David Tamborero, Valtteri Wirta, Richard D Baird, Karen Barker, Fabrice Barlesi, Michael Baumann, Jonas Bergh, Filippo de Braud, Karim Fizazi, Stefan Fröhling, Alejandro Piris-Giménez, Kenneth Seamon, Michiel S Van der Heijden, Wilbert Zwart, Josep Tabernero
Cancer Core Europe brings together the expertise, resources, and interests of seven leading cancer institutes committed to leveraging collective innovation and collaboration in precision oncology. Through targeted efforts addressing key medical challenges in cancer and partnerships with multiple stakeholders, the consortium seeks to advance cancer research and enhance equitable patient care.
{"title":"Cancer Core Europe: Leveraging Institutional Synergies to Advance Oncology Research and Care Globally.","authors":"Javier Carmona, Elena Chavarria, Kate Donoghue, Christina von Gertten, Petra Oberrauch, Emma Pailler, Giovanni Scoazec, Ruud Weijer, Judith Balmaña, Irene Brana, Cinzia Brunelli, Suzette Delaloge, Marc Deloger, Pierre Delpy, Ingemar Ernberg, Rebecca C Fitzgerald, Elena Garralda, Martin Lablans, Janne Lëhtio, Carlos Lopez, Maialen Fernández, Rosalba Miceli, Paolo Nuciforo, Raquel Perez-Lopez, Elena Provenzano, Marjanka K Schmidt, Cesar Serrano, Neeltje Steeghs, David Tamborero, Valtteri Wirta, Richard D Baird, Karen Barker, Fabrice Barlesi, Michael Baumann, Jonas Bergh, Filippo de Braud, Karim Fizazi, Stefan Fröhling, Alejandro Piris-Giménez, Kenneth Seamon, Michiel S Van der Heijden, Wilbert Zwart, Josep Tabernero","doi":"10.1158/2159-8290.CD-24-0377","DOIUrl":"10.1158/2159-8290.CD-24-0377","url":null,"abstract":"<p><p>Cancer Core Europe brings together the expertise, resources, and interests of seven leading cancer institutes committed to leveraging collective innovation and collaboration in precision oncology. Through targeted efforts addressing key medical challenges in cancer and partnerships with multiple stakeholders, the consortium seeks to advance cancer research and enhance equitable patient care.</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":" ","pages":"1147-1153"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141316747","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bone is the most common site of breast cancer metastasis. Bone metastasis is incurable and is associated with severe morbidity. Utilizing an immunocompetent mouse model of spontaneous breast cancer bone metastasis, we profiled the immune transcriptome of bone metastatic lesions and peripheral bone marrow at distinct metastatic stages, revealing dynamic changes during the metastatic process. We show that cross-talk between granulocytes and T cells is central to shaping an immunosuppressive microenvironment. Specifically, we identified the PD-1 and TIGIT signaling axes and the proinflammatory cytokine IL1β as central players in the interactions between granulocytes and T cells. Targeting these pathways in vivo resulted in attenuated bone metastasis and improved survival, by reactivating antitumor immunity. Analysis of patient samples revealed that TIGIT and IL1β are prominent in human bone metastasis. Our findings suggest that cotargeting immunosuppressive granulocytes and dysfunctional T cells may be a promising novel therapeutic strategy to inhibit bone metastasis. Significance: Temporal transcriptome profiling of the immune microenvironment in breast cancer bone metastasis revealed key communication pathways between dysfunctional T cells and myeloid derived suppressor cells. Cotargeting of TIGIT and IL1β inhibited bone metastasis and improved survival. Validation in patient data implicated these targets as a novel promising approach to treat human bone metastasis.
骨是乳腺癌最常见的转移部位。骨转移是无法治愈的,而且与严重的发病率有关。我们利用免疫功能正常的自发性乳腺癌骨转移小鼠模型,分析了骨转移病灶和外周骨髓在不同转移阶段的免疫转录组,揭示了转移过程中的动态变化。我们发现,粒细胞和 T 细胞之间的串扰是形成免疫抑制微环境的核心。具体来说,我们发现 PD-1 和 TIGIT 信号轴以及促炎细胞因子 IL1b 是粒细胞和 T 细胞之间相互作用的核心角色。在体内靶向这些通路可重新激活抗肿瘤免疫,从而减少骨转移并提高生存率。对患者样本的分析表明,TIGIT和IL1b在人类骨转移中表现突出。我们的研究结果表明,联合靶向免疫抑制性粒细胞和功能失调的T细胞可能是一种很有前景的抑制骨转移的新型治疗策略。
{"title":"Combining TIGIT Blockade with MDSC Inhibition Hinders Breast Cancer Bone Metastasis by Activating Antitumor Immunity.","authors":"Lea Monteran, Nour Ershaid, Ye'ela Scharff, Yazeed Zoabi, Tamer Sanalla, Yunfeng Ding, Anna Pavlovsky, Yael Zait, Marva Langer, Tal Caller, Anat Eldar-Boock, Camila Avivi, Amir Sonnenblick, Ronit Satchi-Fainaro, Iris Barshack, Noam Shomron, Xiang H-F Zhang, Neta Erez","doi":"10.1158/2159-8290.CD-23-0762","DOIUrl":"10.1158/2159-8290.CD-23-0762","url":null,"abstract":"<p><p>Bone is the most common site of breast cancer metastasis. Bone metastasis is incurable and is associated with severe morbidity. Utilizing an immunocompetent mouse model of spontaneous breast cancer bone metastasis, we profiled the immune transcriptome of bone metastatic lesions and peripheral bone marrow at distinct metastatic stages, revealing dynamic changes during the metastatic process. We show that cross-talk between granulocytes and T cells is central to shaping an immunosuppressive microenvironment. Specifically, we identified the PD-1 and TIGIT signaling axes and the proinflammatory cytokine IL1β as central players in the interactions between granulocytes and T cells. Targeting these pathways in vivo resulted in attenuated bone metastasis and improved survival, by reactivating antitumor immunity. Analysis of patient samples revealed that TIGIT and IL1β are prominent in human bone metastasis. Our findings suggest that cotargeting immunosuppressive granulocytes and dysfunctional T cells may be a promising novel therapeutic strategy to inhibit bone metastasis. Significance: Temporal transcriptome profiling of the immune microenvironment in breast cancer bone metastasis revealed key communication pathways between dysfunctional T cells and myeloid derived suppressor cells. Cotargeting of TIGIT and IL1β inhibited bone metastasis and improved survival. Validation in patient data implicated these targets as a novel promising approach to treat human bone metastasis.</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":" ","pages":"1252-1275"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140012209","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-24-0612
Aayoung Hong, Gatien Moriceau, Lu Sun, Shirley Lomeli, Marco Piva, Robert Damoiseaux, Sheri L Holmen, Norman E Sharpless, Willy Hugo, Roger S Lo
{"title":"Expression of Concern: Exploiting Drug Addiction Mechanisms to Select against MAPKi-Resistant Melanoma.","authors":"Aayoung Hong, Gatien Moriceau, Lu Sun, Shirley Lomeli, Marco Piva, Robert Damoiseaux, Sheri L Holmen, Norman E Sharpless, Willy Hugo, Roger S Lo","doi":"10.1158/2159-8290.CD-24-0612","DOIUrl":"10.1158/2159-8290.CD-24-0612","url":null,"abstract":"","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":"14 7","pages":"1356"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466375","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-24-0539
Matthew J Hangauer, Jorge Silvio Gutkind, Fleur M Ferguson
In this issue, Ryan and colleagues describe the preclinical development of a pan-RAF:MEK molecular glue with superior efficacy, brain penetrance, and tolerability in xenograft models of Ras/Raf/MAPK pathway-driven tumors. See related article by Ryan et al., p. 1190 (1).
在本期杂志中,Ryan 及其同事介绍了泛 RAF:MEK 分子胶的临床前开发情况,该分子胶在 Ras/Raf/MAPK 通路驱动的肿瘤异种移植模型中具有卓越的疗效、脑穿透性和耐受性。请参见 Ryan 等人的相关文章,第 1190 页(1)。
{"title":"Pan-RAF:MEK Molecular Glues Take Center Stage.","authors":"Matthew J Hangauer, Jorge Silvio Gutkind, Fleur M Ferguson","doi":"10.1158/2159-8290.CD-24-0539","DOIUrl":"10.1158/2159-8290.CD-24-0539","url":null,"abstract":"<p><p>In this issue, Ryan and colleagues describe the preclinical development of a pan-RAF:MEK molecular glue with superior efficacy, brain penetrance, and tolerability in xenograft models of Ras/Raf/MAPK pathway-driven tumors. See related article by Ryan et al., p. 1190 (1).</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":"14 7","pages":"1143-1144"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01DOI: 10.1158/2159-8290.CD-24-0538
Pasquale Lombardi, David J Pinato
Transforming gut microbial status from a prognostic trait to a therapeutic target is a key goal to understand and reverse resistance to anticancer immunotherapy. Glitza and colleagues propose selective manipulation of the gut microbiome with SER401 following antibiotic preconditioning and highlight multiple challenges in delivering microbiome manipulation to the clinic. See related article by Glitza et al., p. 1161 (8).
{"title":"Precision Targeting of the Gut Microbiome for Cancer Immunotherapy.","authors":"Pasquale Lombardi, David J Pinato","doi":"10.1158/2159-8290.CD-24-0538","DOIUrl":"https://doi.org/10.1158/2159-8290.CD-24-0538","url":null,"abstract":"<p><p>Transforming gut microbial status from a prognostic trait to a therapeutic target is a key goal to understand and reverse resistance to anticancer immunotherapy. Glitza and colleagues propose selective manipulation of the gut microbiome with SER401 following antibiotic preconditioning and highlight multiple challenges in delivering microbiome manipulation to the clinic. See related article by Glitza et al., p. 1161 (8).</p>","PeriodicalId":9430,"journal":{"name":"Cancer discovery","volume":"14 7","pages":"1140-1142"},"PeriodicalIF":29.7,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}