Molecular Cancer Therapeutics最新文献

筛选
英文 中文
PROTAC-Mediated Degradation of TAF1 Induces Apoptosis in AML Cells and Inhibits Tumor Growth In Vivo. protac介导的TAF1降解诱导AML细胞凋亡并抑制肿瘤生长
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-16 DOI: 10.1158/1535-7163.MCT-24-1091
Lihong Chen, Zachary P Shultz, Marianna Sansone, Bin Fang, Xiang Liu, Mingxiang Teng, Ernst Schonbrunn, Justin M Lopchuk, Jiandong Chen
{"title":"PROTAC-Mediated Degradation of TAF1 Induces Apoptosis in AML Cells and Inhibits Tumor Growth In Vivo.","authors":"Lihong Chen, Zachary P Shultz, Marianna Sansone, Bin Fang, Xiang Liu, Mingxiang Teng, Ernst Schonbrunn, Justin M Lopchuk, Jiandong Chen","doi":"10.1158/1535-7163.MCT-24-1091","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-1091","url":null,"abstract":"<p><p>The bromodomain-containing protein, transcription factor IID subunit 1 (TAF1; transcription factor II-250), is the largest component of the multiprotein assembly transcription factor IID, a dynamic complex that serves as a general factor for transcription initiation. CRISPR and RNAi screens of pan-cancer cell lines revealed that TAF1 is broadly required for optimal cell growth and survival, but a subset of cell lines showed enhanced TAF1 dependence. These observations suggest that TAF1 has the potential to serve as a therapeutic target in sensitive tumors. Current approaches employed to target TAF1 are limited to monovalent small-molecule inhibitors of the bromodomain. However, recent studies showed that such inhibitors lack cancer cell kill potential. We applied a structure-guided approach to generate cereblon recruiting Proteolysis Targeting Chimera (PROTAC) degraders of TAF1 using the chemical scaffolds of ceralasertib and GNE371. We present evidence that GNE371-based PROTACs are effective in degradation of TAF1 at concentrations as low as 1 nmol/L. TAF1 depletion activated p53 and induced apoptosis in acute myeloid leukemia (AML) cell lines and certain solid tumor cells. An in vivo active TAF1 PROTAC inhibited the growth of AML tumor xenografts. The results showed that inhibition of the bromodomain is not sufficient to inactivate TAF1 functions, whereas a PROTAC approach induces strong biological effects. Furthermore, TAF1 PROTACs have therapeutic potential against AML and other sensitive tumors.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"OF1-OF10"},"PeriodicalIF":5.3,"publicationDate":"2025-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144079141","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 Novel B7-H4xCD3 Bispecific T Cell Engager (PF-07260437) Synergizes with Breast Cancer Standard of Care and Immune-Checkpoint Therapies. 一种新的B7-H4xCD3双特异性T细胞结合剂(PF-07260437)与乳腺癌标准护理和免疫检查点疗法协同作用
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-14 DOI: 10.1158/1535-7163.MCT-24-0379
Keith Abayasiriwardana, Lei Wu, Hanane Laklai, Malgorzata Nocula-Lugowska, Lioudmila Tchistiakova, Jatin Narula, Amy Jackson-Fisher, Jonathan Golas, My-Hanh Lam, Veronika Grinstein, Jung Wook Kang, Jessica C Kearney, Christine Hosselet, Erik Upeslacis, LuAnna Lemon, Yun Zhang, Changhua Ji, Bernard S Buetow, Martin B Finkelstein, Netonia Marshall, Stephanie Bisulco, Edward Rosfjord, Divya Mathur, Jennifer Athanacio, Ashley Thomas, Alexander Trageser, Diane Fernandez, Ziyue Karen Jiang, Sripad Ram, Edward Cabral, Lisa Manzuk, Kevin Maresca, Anand Giddabasappa, Clare Lees, Andrea T Hooper, Puja Sapra, Sudhakar Chintharlapalli
{"title":"A Novel B7-H4xCD3 Bispecific T Cell Engager (PF-07260437) Synergizes with Breast Cancer Standard of Care and Immune-Checkpoint Therapies.","authors":"Keith Abayasiriwardana, Lei Wu, Hanane Laklai, Malgorzata Nocula-Lugowska, Lioudmila Tchistiakova, Jatin Narula, Amy Jackson-Fisher, Jonathan Golas, My-Hanh Lam, Veronika Grinstein, Jung Wook Kang, Jessica C Kearney, Christine Hosselet, Erik Upeslacis, LuAnna Lemon, Yun Zhang, Changhua Ji, Bernard S Buetow, Martin B Finkelstein, Netonia Marshall, Stephanie Bisulco, Edward Rosfjord, Divya Mathur, Jennifer Athanacio, Ashley Thomas, Alexander Trageser, Diane Fernandez, Ziyue Karen Jiang, Sripad Ram, Edward Cabral, Lisa Manzuk, Kevin Maresca, Anand Giddabasappa, Clare Lees, Andrea T Hooper, Puja Sapra, Sudhakar Chintharlapalli","doi":"10.1158/1535-7163.MCT-24-0379","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0379","url":null,"abstract":"<p><p>Immune checkpoint inhibitors have shown limited success in breast cancer, the most common and deadly cancer in women worldwide. Novel immune therapies like CD3 engaging bispecific antibodies have shown clinical promise in hematologic malignancies. However, developing CD3 bispecifics for solid tumors has been challenging due to difficulty in identifying tumor-specific antigens. B7-H4 is proposed as an attractive tumor-associated antigen for breast cancer therapeutics with comprehensive coverage regardless of breast cancer molecular subtype. We designed a B7-H4 targeting CD3 bispecific molecule, PF-07260437, and demonstrated B7-H4-dependent pharmacology in vitro by directing cytotoxic T cell killing to breast cancer cell lines. Treatment of cell line-derived xenograft and patient-derived xenograft in vivo models of human breast cancer with PF-07260437 induced substantial tumoricidal activity, often resulting in complete responses. Mechanistically, PF-07260437 increased T cell number and activation, leading to efficient tumor killing. Additionally, combining PF-07260437 with standard of care (palbociclib plus fulvestrant) and a checkpoint inhibitor (anti-PD-1) showed combinatorial benefits in an immune competent in vivo model. Clinically relevant non-invasive PET/CT imaging with a CD8-targeting tracer demonstrated PF-07260437-mediated increases in intratumoral CD8 T cells highlighting the utility of CD8-PET technology to potentially assess biomarker changes in the clinic. Finally, the manageable toxicity profile of PF-07260437 was highlighted in an exploratory toxicology study in cynomolgus monkeys. These data support the clinical testing of PF-07260437 for treating B7-H4 expressing solid tumors, including breast cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144003210","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
Inhibition of MCL-1 and MEK overcomes MEK inhibitor resistance in triple-negative and inflammatory breast cancers. MCL-1和MEK的抑制克服了三阴性和炎性乳腺癌中MEK抑制剂的耐药性。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-13 DOI: 10.1158/1535-7163.MCT-24-0593
Mohd Mughees, Moises J Tacam, Alex W Tan, Mary K Pitner, LaKesla R Iles, Xiaoding Hu, Emilly S Villodre, Bisrat G Debeb, Takahiro Kogawa, Bora Lim, Rachel M Layman, Wendy A Woodward, Naoto T Ueno, Debu Tripathy, Savitri Krishnamurthy, Yuan Qi, Lajos Pusztai, Jian Wang, Varsha Gandhi, Geoffrey Bartholomeusz, Chandra Bartholomeusz
{"title":"Inhibition of MCL-1 and MEK overcomes MEK inhibitor resistance in triple-negative and inflammatory breast cancers.","authors":"Mohd Mughees, Moises J Tacam, Alex W Tan, Mary K Pitner, LaKesla R Iles, Xiaoding Hu, Emilly S Villodre, Bisrat G Debeb, Takahiro Kogawa, Bora Lim, Rachel M Layman, Wendy A Woodward, Naoto T Ueno, Debu Tripathy, Savitri Krishnamurthy, Yuan Qi, Lajos Pusztai, Jian Wang, Varsha Gandhi, Geoffrey Bartholomeusz, Chandra Bartholomeusz","doi":"10.1158/1535-7163.MCT-24-0593","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0593","url":null,"abstract":"<p><p>The MAPK pathway can drive resistance in highly aggressive breast cancers. Our previous work showed that MEK inhibitor (MEKi) AZD6244 (selumetinib) prevented lung metastasis in a breast cancer xenograft model. In clinical studies, MEKis as single agents have had only modest activity against solid tumors due to the onset of resistance. Using synthetic-lethality siRNA screening, we identified myeloid cell leukemia-1 (MCL-1) as a potential contributor to AZD6244 resistance. We hypothesized that MCL-1 promotes MEKi resistance in highly aggressive breast cancers and that MCL-1 inhibition overcomes AZD6244 resistance. We established two AZD6244-resistant cell lines: MDA-MB-231-R (triple-negative breast cancer) and SUM149-R (triple-negative inflammatory breast cancer). These resistant cells were characterized with respect to different parameters, and a combination of an MCL-1 inhibitor (MCL-1i) together with a MEKi was evaluated in vitro and in vivo to overcome the acquired resistance. Compared with their respective parental cells, MDA-MB-231-R and SUM149-R cells showed increased proliferation, colony formation, stemness, anchorage-independent growth, and MCL-1 expression levels. MCL-1 knockdown in resistant cells decreased cell proliferation and colony formation, increased apoptosis, and was associated with high expression of the pro-apoptotic proteins PUMA, NOXA, BAK, and BAX. MEKi resistance was overcome when resistant cells were treated with MCL-1i and MEKi combined. In an in vivo mouse model, inhibition of MCL-1 restored sensitivity to AZD6244. Our results suggest that MCL-1 is a driver of MEKi resistance and that combining an MCL-1i with a MEKi warrants further investigation in triple-negative and triple-negative inflammatory breast cancer.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143979126","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
Characterization of mAb104, a Monoclonal Antibody Targeting a Conformationally Exposed, Tumor-specific epitope of HER2. 靶向HER2构象暴露的肿瘤特异性表位的单克隆抗体mAb104的鉴定
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-13 DOI: 10.1158/1535-7163.MCT-24-0583
Sagun Parakh, Nhi Huynh, Diana Dong Cao, Angela Rigopoulos, Benjamin Gloria, Ingrid J G Burvenich, Carmel Murone, Christian W Wichmann, Nancy Yanan Guo, Clare Senko, Adam Parslow, Laura Allan, Laura D Osellame, Peter W Janes, Fiona E Scott, Zhanqi Liu, Hui K Gan, Andrew M Scott
{"title":"Characterization of mAb104, a Monoclonal Antibody Targeting a Conformationally Exposed, Tumor-specific epitope of HER2.","authors":"Sagun Parakh, Nhi Huynh, Diana Dong Cao, Angela Rigopoulos, Benjamin Gloria, Ingrid J G Burvenich, Carmel Murone, Christian W Wichmann, Nancy Yanan Guo, Clare Senko, Adam Parslow, Laura Allan, Laura D Osellame, Peter W Janes, Fiona E Scott, Zhanqi Liu, Hui K Gan, Andrew M Scott","doi":"10.1158/1535-7163.MCT-24-0583","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0583","url":null,"abstract":"<p><p>We generated a novel HER2 monoclonal antibody (mAb104) which binds to an epitope in domain II of HER2 that is conformationally exposed in tumors in response to HER2 amplification or activation but is not accessible to antibody binding in normal tissues. Consistent with other studies that evaluated antibodies targeting conformationally exposed epitopes, mAb104 lacked in vitro activity however showed potent anti-tumor activity in vivo. The anti-tumor effect in vivo was similar in magnitude to trastuzumab and pertuzumab, whilstand combination with trastuzumab was superior to trastuzumab alone. Immunohistochemistry screening of normal and tumor tissues with mAb104 showed mAb104 did not bind to normal tissues, confirming tumor specificity of mAb104. In vivo biodistribution and imaging data demonstrated specific tumor targeting of mAb104 in HER2 expressing tumors. Confocal microscopy clearly demonstrated internalization of mAb104 into the tumor cells, consistent with mAb104:HER2 trafficking. Mab104 is tumor specific, exhibits potent antitumor activity in HER2-positive models and internalizes into HER2-positive tumor cells. These results demonstrate the potential of mAb104 as a novel HER2 targeting therapy both as a naked antibody for signalling abrogation therapy, and for payload delivery as an antibody-drug conjugate or for beta/alpha particle therapy.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144018164","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
CRISPR screens identify POLB as a synthetic lethal enhancer of PARP inhibition exclusively in BRCA-mutated tumors. CRISPR筛选鉴定出POLB是仅在brca突变肿瘤中抑制PARP的合成致死增强子。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-06 DOI: 10.1158/1535-7163.MCT-24-0822
Katherine Lazarides, Justin L Engel, Michele Meseonznik, Tianshu Feng, Ashley H Choi, Yi Yu, Shangtao Liu, Samuel R Meier, Hongxiang Zhang, Binzhang Shen, Robert Tjin Tham Sjin, Douglas A Whittington, Brian J McMillan, Brian Doyon, Xuewen Pan, Erik Wilker, Alan Huang, Jannik N Andersen, William D Mallender, Madhavi Bandi
{"title":"CRISPR screens identify POLB as a synthetic lethal enhancer of PARP inhibition exclusively in BRCA-mutated tumors.","authors":"Katherine Lazarides, Justin L Engel, Michele Meseonznik, Tianshu Feng, Ashley H Choi, Yi Yu, Shangtao Liu, Samuel R Meier, Hongxiang Zhang, Binzhang Shen, Robert Tjin Tham Sjin, Douglas A Whittington, Brian J McMillan, Brian Doyon, Xuewen Pan, Erik Wilker, Alan Huang, Jannik N Andersen, William D Mallender, Madhavi Bandi","doi":"10.1158/1535-7163.MCT-24-0822","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0822","url":null,"abstract":"<p><p>Poly (ADP-ribose) polymerases inhibitors (PARPi) are an approved class of anticancer therapeutics that inhibit the activities of PARP1/2 and produce synthetic lethality in BRCA1/2 mutated cancers due to the absence of a functional homologous recombination (HR)-dependent DNA repair pathway. Although PARP inhibitors have led to successful clinical outcomes, two-thirds of patients develop acquired resistance, limiting long-term utility as maintenance therapy. Motivated by this clinical need, we utilized a CRISPR target discovery screening platform to identify POLB as a gene that acts selectively and synergistically with PARPi in BRCA1/2 mutated cancers and found that POLB knockout along with PARPi treatment enhanced loss of viability in BRCA1/2 mutant and BRCA2 null cells, but not in isogenic BRCA1/2 wildtype cells. Overexpression of either POLB wildtype or catalytically inactive mutants confirmed that perturbation of both the polymerase and lyase catalytic activities of POLB are required for synergistic PARP-BRCA synthetic lethality. Mechanistically, POLB knockout was associated with an increase in single- and double-strand DNA breaks, cell cycle arrest, and apoptosis when in combination with PARP inhibition. The translational nature of this interaction was further examined using murine xenograft models of BRCA1 mutant and BRCA2 null cell lines, wherein the combination of POLB knockout and niraparib led to profound tumor regression and prevented tumor regrowth even after cessation of treatment. Together, these results suggest that POLB is a synergistic enhancer of the synthetic lethal interaction between PARP and BRCA and support POLB as a promising therapeutic target for improving antitumor responses to PARPi in HR-deficient cancers.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-05-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144024970","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
Novel chimeric CTLA-4 B-cell epitope peptide vaccines demonstrate effective anti-tumor immunity with/without PD1/PDL1 blockade in multiple syngeneic murine models of breast and colorectal cancers. 新型嵌合CTLA-4 b细胞表位肽疫苗在具有或不具有PD1/PDL1阻断的多种同基因小鼠乳腺癌和结直肠癌模型中显示出有效的抗肿瘤免疫。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-06 DOI: 10.1158/1535-7163.MCT-24-0908
Linlin Guo, Jay Overholser, Sarah Naylon, Stephane Roche, Nicholas Ede, Pravin T P Kaumaya
{"title":"Novel chimeric CTLA-4 B-cell epitope peptide vaccines demonstrate effective anti-tumor immunity with/without PD1/PDL1 blockade in multiple syngeneic murine models of breast and colorectal cancers.","authors":"Linlin Guo, Jay Overholser, Sarah Naylon, Stephane Roche, Nicholas Ede, Pravin T P Kaumaya","doi":"10.1158/1535-7163.MCT-24-0908","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-24-0908","url":null,"abstract":"<p><p>Cancer immunotherapy with checkpoint inhibitors has resulted in impressive clinical results in several cancer indications. Despite this success, only a fraction of patients shows durable and complete response to blockade by Ipilimumab an anti-CTLA-4 monoclonal antibody. We identified four cytotoxic T Lymphocyte-associated antigen 4 (CTLA-4) peptide sequences from which engineered chimeric B-cell epitope constructs incorporating a \"promiscuous\" T-cell epitope elicited highly immunogenic anti-CTLA-4 natural polyclonal antibodies by immunization. Combination of CTLA-4 peptide vaccine with other checkpoint inhibitor vaccines PD1-Vaxx or PDL1-Vaxx were investigated in several breast and colon carcinoma BALB/c syngeneic models (CT26, 4T1 and D2F2). CTLA-4 vaccines showed significant tumor suppression and prolonged survival rates as compared to anti-mouse CTLA-4 mAb 9H10. The resulting antipeptide antibodies suppressed tumor proliferation and migration similar to ipilimumab. We focused on one CTLA-4 epitope sequence 130-150 that embodies the \"MYPPPY\" motif that ipilimumab binds to. Combination of MVF-CTLA-4(130-150) with either PD1-Vaxx or PDL1-Vaxx showed synergistic activity. The 130-150 peptide mimic demonstrated the polyproline type II helix motif showed inhibition of tumor growth and therapeutic efficacy in the syngeneic CT26/BALB/c model. Several CTLA-4 vaccines have been identified which shows synergistic activities with other checkpoint inhibitor vaccines to PD-1 and PDL1.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-05-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144001847","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
Inhibition of DKK-1 by WAY262611 Inhibits Osteosarcoma Metastasis. WAY262611抑制DKK-1抑制骨肉瘤转移
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-02 DOI: 10.1158/1535-7163.MCT-24-0744
Adit Tal, Shimara Gunawardana-Zeigler, Da Peng, Yuqi Tan, Natalia Munoz Perez, Rachel Offenbacher, Laurel Kastner, Paul Ciero, Matthew E Randolph, Yun Gong, Hong-Wen Deng, Patrick Cahan, David M Loeb
{"title":"Inhibition of DKK-1 by WAY262611 Inhibits Osteosarcoma Metastasis.","authors":"Adit Tal, Shimara Gunawardana-Zeigler, Da Peng, Yuqi Tan, Natalia Munoz Perez, Rachel Offenbacher, Laurel Kastner, Paul Ciero, Matthew E Randolph, Yun Gong, Hong-Wen Deng, Patrick Cahan, David M Loeb","doi":"10.1158/1535-7163.MCT-24-0744","DOIUrl":"10.1158/1535-7163.MCT-24-0744","url":null,"abstract":"<p><p>Osteosarcoma is the most common primary malignant bone tumor in childhood. Patients who present with metastatic disease at diagnosis or relapse have a very poor prognosis, and this has not changed over the past four decades. The Wnt signaling pathway plays a role in regulating osteogenesis and is implicated in osteosarcoma pathogenesis. DKK-1 inhibits the canonical Wnt signaling pathway, causing inhibition of osteoblast differentiation and disordered bone repair. Our lab previously demonstrated that an mAb against DKK-1 prevented metastatic disease in a mouse model. This study expands upon those findings by demonstrating similar results with a small-molecule inhibitor of DKK-1, WAY262611, both in vitro and in vivo. WAY262611 was evaluated in vitro on osteosarcoma cell lines, including proliferation, caspase activation, cell-cycle analysis, and signaling pathway activation. We utilized our orthotopic implantation/amputation model of osteosarcoma metastasis in vivo to determine the impact of WAY262611 on primary tumor progression and metastatic outgrowth of disseminated tumor cells. Differentiation status was determined using single-cell RNA sequencing. We show here that WAY262611 activates canonical Wnt signaling, enhances nuclear localization and transcriptional activity of β-catenin, and slows proliferation of osteosarcoma cell lines. We also show that WAY262611 induces osteoblastic differentiation of a patient-derived xenograft of osteosarcoma in vivo, as well as inhibiting metastasis. This work credentials DKK-1 as a therapeutic target in osteosarcoma, allowing for manipulation of the Wnt signaling pathway and providing preclinical justification for the development of new biologics for the prevention of osteosarcoma metastasis.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"728-739"},"PeriodicalIF":5.3,"publicationDate":"2025-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12048250/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142951624","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
Dual-Nuclide Biodistribution and Therapeutic Evaluation of a Novel Antibody-Based Radiopharmaceutical in Anaplastic Thyroid Cancer Xenografts. 一种新的基于抗体的放射性药物在间变性甲状腺癌异种移植中的双核素生物分布和治疗评价。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-02 DOI: 10.1158/1535-7163.MCT-24-0524
Anja Charlotte Lundgren Mortensen, Hanna Berglund, Preeti Jha, Adam Stenman, Ram Kumar Selvaraju, Hans Lundqvist, Camilla Hofström, Helena Persson, C Christofer Juhlin, Jan Zedenius, Fredrik Y Frejd, Marika Nestor
{"title":"Dual-Nuclide Biodistribution and Therapeutic Evaluation of a Novel Antibody-Based Radiopharmaceutical in Anaplastic Thyroid Cancer Xenografts.","authors":"Anja Charlotte Lundgren Mortensen, Hanna Berglund, Preeti Jha, Adam Stenman, Ram Kumar Selvaraju, Hans Lundqvist, Camilla Hofström, Helena Persson, C Christofer Juhlin, Jan Zedenius, Fredrik Y Frejd, Marika Nestor","doi":"10.1158/1535-7163.MCT-24-0524","DOIUrl":"10.1158/1535-7163.MCT-24-0524","url":null,"abstract":"<p><p>Anaplastic thyroid cancer (ATC) is a rare but severe form of thyroid cancer responsible for approximately 50% of thyroid cancer deaths. Consequently, the identification of innovative therapies remains crucial for the effective treatment of ATC. Molecular radiotherapy is a rapidly growing field within oncology, and the cell surface antigen CD44v6, which is overexpressed in several cancers, is a plausible target for molecular radiotherapy of ATC. IHC of 39 patient samples with ATC was evaluated for CD44v6 expression. Biodistribution and dosimetry of iodine-125 (125I)-/lutetium-177 (177Lu)-labeled UU-40, a CD44v6-specific antibody, followed by in vivo efficacy in two ATC xenograft models with varying target expression levels (ACT-1 and BHT-101), accompanied by single-photon emission computed tomography (SPECT) imaging, evaluated radiolabeled UU-40 for therapeutic efficiency in ATC xenografts. The IHC revealed CD44v6 immunoreactivity in 46% of patient samples with ATC. The biodistribution favored 177Lu-labeled UU-40 over the 125I-labeled antibody and confirmed the in vivo specificity of both radioconjugates. The in vivo efficacy and accompanied SPECT imaging of a moderate CD44v6-expressing xenograft model (BHT-101) verified the tumor specificity, as well as the target-specific effect of 177Lu-labeled UU-40 on tumor growth and survival. A 100% complete response rate was demonstrated as a result of therapy using a single dose of 16 MBq 177Lu-labeled UU-40 in a high CD44v6-expressing xenograft model (ACT-1), and SPECT imaging revealed excellent tumor uptake of the radioconjugate at 14 days after injection. This study verifies the expression of CD44v6 in ATC and strengthens the superiority and promise of 177Lu-labeled UU-40 over 131I-labeled UU-40 for antibody-based molecular radiotherapy of CD44v6-positive ATC.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"753-762"},"PeriodicalIF":5.3,"publicationDate":"2025-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12046332/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143458675","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
A Vision for the Future of Molecular Cancer Therapeutics. 展望分子癌症治疗的未来。
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-02 DOI: 10.1158/1535-7163.MCT-25-0350
Daniel K Nomura
{"title":"A Vision for the Future of Molecular Cancer Therapeutics.","authors":"Daniel K Nomura","doi":"10.1158/1535-7163.MCT-25-0350","DOIUrl":"https://doi.org/10.1158/1535-7163.MCT-25-0350","url":null,"abstract":"","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":"24 5","pages":"663"},"PeriodicalIF":5.3,"publicationDate":"2025-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143970829","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
First-Line Apatinib Combined with Tislelizumab and Chemotherapy for Patients with Advanced Gastric and Gastroesophageal Junction Adenocarcinoma with Poor Prognosis. 一线阿帕替尼联合替利单抗和化疗治疗预后不良的晚期胃和胃食管交界处腺癌患者
IF 5.3 2区 医学
Molecular Cancer Therapeutics Pub Date : 2025-05-02 DOI: 10.1158/1535-7163.MCT-24-0143
Wenjie Zhang, Jing Wang, Wei Shi, Hu Qiu, Shaobo Ke, Yuanyuan Tian, Yi Gong, Caiyutian Zhang, Jiamei Chen, Yong Wu, Wensi Zhao, Yongshun Chen
{"title":"First-Line Apatinib Combined with Tislelizumab and Chemotherapy for Patients with Advanced Gastric and Gastroesophageal Junction Adenocarcinoma with Poor Prognosis.","authors":"Wenjie Zhang, Jing Wang, Wei Shi, Hu Qiu, Shaobo Ke, Yuanyuan Tian, Yi Gong, Caiyutian Zhang, Jiamei Chen, Yong Wu, Wensi Zhao, Yongshun Chen","doi":"10.1158/1535-7163.MCT-24-0143","DOIUrl":"10.1158/1535-7163.MCT-24-0143","url":null,"abstract":"<p><p>In this prospective, open-label, exploratory study (RENMIN-213), patients with previously untreated, HER2-negative, advanced G/GEJ adenocarcinoma with signet ring cell carcinoma or peritoneal metastasis were enrolled to receive eight cycles of apatinib, tislelizumab, and chemotherapy every 3 weeks, with a maintenance therapy with apatinib plus tislelizumab for a maximum of 1 year. Homogeneous patients receiving immune checkpoint inhibitors combined with chemotherapy at the same time were deemed as the control group for efficacy. The primary endpoint was progression-free survival. Secondary endpoints were objective response rate, disease control rate, duration of response, overall survival, biomarkers, health-related quality of life, and safety. A total of 33 patients [median (range) age, 60 (32-72) years; 21 (63.6%) male; 11 (33.3%) signet ring cell carcinoma; 30 (90.9%) peritoneal metastasis] were enrolled and deemed evaluable for efficacy analysis. Of these patients, 32 (97%) were without disease progression, of whom 1 (3.0%) patient had complete response and 18 (54.6%) had partial response. Six patients (18.2%) were able to undergo surgery after treatment. After propensity score matching, the median progression-free survival was 10.17 months (95% confidence interval, 7.13-13.21) in the apatinib combined with tislelizumab and chemotherapy group, as compared with 6.0 months in the immune checkpoint inhibitor combined with chemotherapy group. The median duration of response increased from 4.5 to 8.7 months. The objective response rate increased from 33.3% to 54.6%, and the disease control rate increased from 87.9% to 97.0%. Treatment-related grade 3 or higher adverse events for evaluable patients occurred in 11 patients (33.3%), and patients' health-related quality of life improved after treatment.</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":" ","pages":"720-727"},"PeriodicalIF":5.3,"publicationDate":"2025-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143080516","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信