Pub Date : 2024-07-19DOI: 10.18632/oncotarget.28615
Caroline Naomi Valdez, Gabriela Athziri Sánchez-Zuno, Lais Osmani, Wael Ibrahim, Anjela Galan, Antonietta Bacchiocchi, Ruth Halaban, Rajan P Kulkarni, Insoo Kang, Richard Bucala, Thuy Tran
Macrophage Migration Inhibitory Factor (MIF) and its homolog D-dopachrome Tautomerase (DDT) have been implicated as drivers of tumor progression across a variety of cancers. Recent evidence suggests MIF as a therapeutic target in immune checkpoint inhibition (ICI) resistant melanomas, however clinical evidence of MIF and particularly of DDT remain limited. This retrospective study analyzed 97 patients treated at Yale for melanoma between 2002-2020. Bulk-RNA sequencing of patient tumor samples from the Skin Cancer SPORE Biorepository was used to evaluate for differential gene expression of MIF, DDT, CD74, and selected inflammatory markers, and gene expression was correlated with patient survival outcomes. Our findings revealed a strong correlation between MIF and DDT levels, with no statistically significant difference across common melanoma mutations and subtypes. Improved survival was associated with lower MIF and DDT levels and higher CD74:MIF and CD74:DDT levels. High CD74:DDT and CD74:MIF levels were also associated with enrichment of infiltrating inflammatory cell markers. These data suggest DDT as a novel target in immune therapy. Dual MIF and DDT blockade may provide synergistic responses in patients with melanoma, irrespective of common mutations, and may overcome ICI resistance. These markers may also provide prognostic value for further biomarker development.
{"title":"Prognostic and therapeutic insights into MIF, DDT, and CD74 in melanoma.","authors":"Caroline Naomi Valdez, Gabriela Athziri Sánchez-Zuno, Lais Osmani, Wael Ibrahim, Anjela Galan, Antonietta Bacchiocchi, Ruth Halaban, Rajan P Kulkarni, Insoo Kang, Richard Bucala, Thuy Tran","doi":"10.18632/oncotarget.28615","DOIUrl":"10.18632/oncotarget.28615","url":null,"abstract":"<p><p>Macrophage Migration Inhibitory Factor (MIF) and its homolog D-dopachrome Tautomerase (DDT) have been implicated as drivers of tumor progression across a variety of cancers. Recent evidence suggests MIF as a therapeutic target in immune checkpoint inhibition (ICI) resistant melanomas, however clinical evidence of MIF and particularly of DDT remain limited. This retrospective study analyzed 97 patients treated at Yale for melanoma between 2002-2020. Bulk-RNA sequencing of patient tumor samples from the Skin Cancer SPORE Biorepository was used to evaluate for differential gene expression of MIF, DDT, CD74, and selected inflammatory markers, and gene expression was correlated with patient survival outcomes. Our findings revealed a strong correlation between MIF and DDT levels, with no statistically significant difference across common melanoma mutations and subtypes. Improved survival was associated with lower MIF and DDT levels and higher CD74:MIF and CD74:DDT levels. High CD74:DDT and CD74:MIF levels were also associated with enrichment of infiltrating inflammatory cell markers. These data suggest DDT as a novel target in immune therapy. Dual MIF and DDT blockade may provide synergistic responses in patients with melanoma, irrespective of common mutations, and may overcome ICI resistance. These markers may also provide prognostic value for further biomarker development.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"507-520"},"PeriodicalIF":0.0,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11259151/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141724106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-17DOI: 10.18632/oncotarget.28527
Yiyao Zhang, Li Liu, Pei Fan, Nathalie Bauer, Jury Gladkich, Eduard Ryschich, Alexandr V Bazhin, Nathalia A Giese, Oliver Strobel, Thilo Hackert, Ulf Hinz, Wolfgang Gross, Franco Fortunato, Ingrid Herr
{"title":"Correction: Aspirin counteracts cancer stem cell features, desmoplasia and gemcitabine resistance in pancreatic cancer.","authors":"Yiyao Zhang, Li Liu, Pei Fan, Nathalie Bauer, Jury Gladkich, Eduard Ryschich, Alexandr V Bazhin, Nathalia A Giese, Oliver Strobel, Thilo Hackert, Ulf Hinz, Wolfgang Gross, Franco Fortunato, Ingrid Herr","doi":"10.18632/oncotarget.28527","DOIUrl":"10.18632/oncotarget.28527","url":null,"abstract":"","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"504-506"},"PeriodicalIF":0.0,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11254298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141634144","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-17DOI: 10.18632/oncotarget.28606
Kseniya Sinichenkova, Iliya Sidorov, Nataliya Kriventsova, Dmitriy Konovalov, Ruslan Abasov, Nataliya Usman, Alexander Karachunskiy, Galina Novichkova, Dmitriy Litvinov, Alexander Druy
Introduction: BRAF V600E substitution predicts sensitivity of a cancer to BRAF inhibitor therapy. The mutation is rarely found in soft-tissue sarcomas. Here we describe a case of undifferentiated spindle cell sarcoma showing primary insensitivity to standard chemotherapy and pronounced but non-sustained response to BRAF/MEK inhibitors at recurrence.
Case presentation: A 13-year-old girl was diagnosed with low-grade spindle cell sarcoma of pelvic localization, BRAF exon 15 double-mutated: c.1799T>A p.V600E and c.1819T>A p.S607T in cis-position. The tumor showed resistance to CWS-based first-line chemotherapy and was treated surgically by radical resection. Seven months after surgery the patient developed metastatic relapse with abdominal carcinomatosis. Combined targeted therapy with BRAF/MEK inhibitors afforded complete response in 1 month and was continued, though complicated by severe side effects (fever, rash) necessitating 1-2 week toxicity breaks. After 4 months from commencement the disease recurred and anti-BRAF/MEK regimen consolidation was unsuccessful. Intensive salvation chemotherapy was ineffective. Empirical immunotherapy afforded a transient partial response giving way to fatal progression with massive, abdominal cocoon-complicated peritoneal carcinomatosis.
Conclusion: This is the first report of spindle cell sarcoma BRAF V600E/S607T double-mutated, responding to a combination of B-Raf and MEK inhibitors. Despite the low histological grade and radical surgical treatment of the tumor at primary manifestation, the disease had aggressive clinical course and the response to BRAF/MEK targeted therapy at recurrence was complete but nondurable. Empirical use of pembrolizumab provided no unambiguous evidence on the clinical relevance of immunotherapy in protein kinase -rearranged spindle cell tumors.
{"title":"Rapid but nondurable response of a <i>BRAF</i> exon 15 double-mutated spindle cell sarcoma to a combination of BRAF and MEK inhibitors.","authors":"Kseniya Sinichenkova, Iliya Sidorov, Nataliya Kriventsova, Dmitriy Konovalov, Ruslan Abasov, Nataliya Usman, Alexander Karachunskiy, Galina Novichkova, Dmitriy Litvinov, Alexander Druy","doi":"10.18632/oncotarget.28606","DOIUrl":"10.18632/oncotarget.28606","url":null,"abstract":"<p><strong>Introduction: </strong>BRAF V600E substitution predicts sensitivity of a cancer to BRAF inhibitor therapy. The mutation is rarely found in soft-tissue sarcomas. Here we describe a case of undifferentiated spindle cell sarcoma showing primary insensitivity to standard chemotherapy and pronounced but non-sustained response to BRAF/MEK inhibitors at recurrence.</p><p><strong>Case presentation: </strong>A 13-year-old girl was diagnosed with low-grade spindle cell sarcoma of pelvic localization, BRAF exon 15 double-mutated: c.1799T>A p.V600E and c.1819T>A p.S607T in cis-position. The tumor showed resistance to CWS-based first-line chemotherapy and was treated surgically by radical resection. Seven months after surgery the patient developed metastatic relapse with abdominal carcinomatosis. Combined targeted therapy with BRAF/MEK inhibitors afforded complete response in 1 month and was continued, though complicated by severe side effects (fever, rash) necessitating 1-2 week toxicity breaks. After 4 months from commencement the disease recurred and anti-BRAF/MEK regimen consolidation was unsuccessful. Intensive salvation chemotherapy was ineffective. Empirical immunotherapy afforded a transient partial response giving way to fatal progression with massive, abdominal cocoon-complicated peritoneal carcinomatosis.</p><p><strong>Conclusion: </strong>This is the first report of spindle cell sarcoma BRAF V600E/S607T double-mutated, responding to a combination of B-Raf and MEK inhibitors. Despite the low histological grade and radical surgical treatment of the tumor at primary manifestation, the disease had aggressive clinical course and the response to BRAF/MEK targeted therapy at recurrence was complete but nondurable. Empirical use of pembrolizumab provided no unambiguous evidence on the clinical relevance of immunotherapy in protein kinase -rearranged spindle cell tumors.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"493-500"},"PeriodicalIF":0.0,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11254299/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141634145","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-17DOI: 10.18632/oncotarget.28610
Jack G Fisher, Lara V Graham, Matthew D Blunt
{"title":"Strategies to disrupt NKG2A:HLA-E interactions for improved anti-cancer immunity.","authors":"Jack G Fisher, Lara V Graham, Matthew D Blunt","doi":"10.18632/oncotarget.28610","DOIUrl":"10.18632/oncotarget.28610","url":null,"abstract":"","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"501-503"},"PeriodicalIF":0.0,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11254306/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141634146","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-16DOI: 10.18632/oncotarget.28612
Eamon Toye, Alexander Chehrazi-Raffle, Justin Hwang, Emmanuel S Antonarakis
Activating mutations in the mitogen-activated protein kinase (MAPK) pathway represent driver alterations governing tumorigenesis, metastasis, and therapy resistance. MAPK activation predominantly occurs through genomic alterations in RAS and BRAF. BRAF is an effector kinase that functions downstream of RAS and propagates this oncogenic activity through MEK and ERK. Across cancers, BRAF alterations include gain-of-function mutations, copy-number alterations, and structural rearrangements. In cancer patients, BRAF-targeting precision therapeutics are effective against Class I BRAF alterations (p.V600 hotspot mutations) in tumors such as melanomas, thyroid cancers, and colorectal cancers. However, numerous non-Class I BRAF inhibitors are also in development and have been explored in some cancers. Here we discuss the diverse forms of BRAF alterations found in human cancers and the strategies to inhibit them in patients harboring cancers of distinct origins.
{"title":"Targeting the multifaceted BRAF in cancer: New directions.","authors":"Eamon Toye, Alexander Chehrazi-Raffle, Justin Hwang, Emmanuel S Antonarakis","doi":"10.18632/oncotarget.28612","DOIUrl":"10.18632/oncotarget.28612","url":null,"abstract":"<p><p>Activating mutations in the mitogen-activated protein kinase (MAPK) pathway represent driver alterations governing tumorigenesis, metastasis, and therapy resistance. MAPK activation predominantly occurs through genomic alterations in <i>RAS</i> and <i>BRAF</i>. BRAF is an effector kinase that functions downstream of <i>RAS</i> and propagates this oncogenic activity through MEK and ERK. Across cancers, <i>BRAF</i> alterations include gain-of-function mutations, copy-number alterations, and structural rearrangements. In cancer patients, BRAF-targeting precision therapeutics are effective against Class I <i>BRAF</i> alterations (p.V600 hotspot mutations) in tumors such as melanomas, thyroid cancers, and colorectal cancers. However, numerous non-Class I BRAF inhibitors are also in development and have been explored in some cancers. Here we discuss the diverse forms of <i>BRAF</i> alterations found in human cancers and the strategies to inhibit them in patients harboring cancers of distinct origins.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"486-492"},"PeriodicalIF":0.0,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11254297/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141634147","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-12DOI: 10.18632/oncotarget.28608
Chantal L Duurland, Thijs de Gunst, Harm C den Boer, Marion T J van den Bosch, Bryony J Telford, Rogier M Vos, Xiaolei Xie, Mingfa Zang, Fang Wang, Yingying Shao, Xiaoyu An, Jingjing Wang, Jie Cai, Ludovic Bourré, Laurens A H van Pinxteren, Roel Q J Schaapveld, Michel Janicot, Sanaz Yahyanejad
microRNAs (miRNAs) are small, non-coding RNAs that regulate expression of multiple genes. MiR-193a-3p functions as a tumor suppressor in many cancer types, but its effect on inducing specific anti-tumor immune responses is unclear. Therefore, we examined the effect of our lipid nanoparticle (LNP) formulated, chemically modified, synthetic miR-193a-3p mimic (INT-1B3) on anti-tumor immunity. INT-1B3 inhibited distant tumor metastasis and significantly prolonged survival. INT-1B3-treated animals were fully protected against challenge with autologous tumor cells even in absence of treatment indicating long-term immunization. Protection against autologous tumor cell challenge was hampered upon T cell depletion and adoptive T cell transfer abrogated tumor growth. Transfection of tumor cells with our miR-193a-3p mimic (1B3) resulted in tumor cell death and apoptosis accompanied by increased expression of DAMPs. Co-culture of 1B3-transfected tumor cells and immature DC led to DC maturation and these mature DC were able to stimulate production of type 1 cytokines by CD4+ and CD8+ T cells. CD4-CD8- T cells also produced type 1 cytokines, even in response to 1B3-transfected tumor cells directly. Live cell imaging demonstrated PBMC-mediated cytotoxicity against 1B3-transfected tumor cells. These data demonstrate for the first time that miR-193a-3p induces long-term immunity against tumor development via modulation of the tumor microenvironment and induction of immunogenic cell death.
微小RNA(miRNA)是一种小型非编码RNA,可调控多个基因的表达。MiR-193a-3p 在许多癌症类型中发挥着肿瘤抑制因子的作用,但其诱导特异性抗肿瘤免疫反应的效果尚不清楚。因此,我们研究了脂质纳米粒子(LNP)配制的化学修饰合成 miR-193a-3p 模拟物(INT-1B3)对抗肿瘤免疫的影响。INT-1B3 可抑制肿瘤远处转移并显著延长生存期。INT-1B3处理过的动物即使在未接受治疗的情况下也能完全抵御自体肿瘤细胞的挑战,这表明INT-1B3具有长期免疫作用。对自体肿瘤细胞挑战的保护在 T 细胞耗竭后受到阻碍,而采用性 T 细胞转移则会抑制肿瘤生长。用我们的 miR-193a-3p mimic(1B3)转染肿瘤细胞会导致肿瘤细胞死亡和凋亡,同时增加 DAMPs 的表达。转染了 1B3 的肿瘤细胞与未成熟的 DC 共同培养会导致 DC 成熟,这些成熟的 DC 能够刺激 CD4+ 和 CD8+ T 细胞产生 1 型细胞因子。CD4-CD8- T细胞也会产生1型细胞因子,甚至直接对1B3转染的肿瘤细胞产生反应。活细胞成像显示了 PBMC 介导的针对 1B3 转染肿瘤细胞的细胞毒性。这些数据首次证明,miR-193a-3p 可通过调节肿瘤微环境和诱导免疫原性细胞死亡,诱导长期的肿瘤免疫。
{"title":"INT-1B3, an LNP formulated miR-193a-3p mimic, promotes anti-tumor immunity by enhancing T cell mediated immune responses via modulation of the tumor microenvironment and induction of immunogenic cell death.","authors":"Chantal L Duurland, Thijs de Gunst, Harm C den Boer, Marion T J van den Bosch, Bryony J Telford, Rogier M Vos, Xiaolei Xie, Mingfa Zang, Fang Wang, Yingying Shao, Xiaoyu An, Jingjing Wang, Jie Cai, Ludovic Bourré, Laurens A H van Pinxteren, Roel Q J Schaapveld, Michel Janicot, Sanaz Yahyanejad","doi":"10.18632/oncotarget.28608","DOIUrl":"10.18632/oncotarget.28608","url":null,"abstract":"<p><p>microRNAs (miRNAs) are small, non-coding RNAs that regulate expression of multiple genes. MiR-193a-3p functions as a tumor suppressor in many cancer types, but its effect on inducing specific anti-tumor immune responses is unclear. Therefore, we examined the effect of our lipid nanoparticle (LNP) formulated, chemically modified, synthetic miR-193a-3p mimic (INT-1B3) on anti-tumor immunity. INT-1B3 inhibited distant tumor metastasis and significantly prolonged survival. INT-1B3-treated animals were fully protected against challenge with autologous tumor cells even in absence of treatment indicating long-term immunization. Protection against autologous tumor cell challenge was hampered upon T cell depletion and adoptive T cell transfer abrogated tumor growth. Transfection of tumor cells with our miR-193a-3p mimic (1B3) resulted in tumor cell death and apoptosis accompanied by increased expression of DAMPs. Co-culture of 1B3-transfected tumor cells and immature DC led to DC maturation and these mature DC were able to stimulate production of type 1 cytokines by CD4+ and CD8+ T cells. CD4-CD8- T cells also produced type 1 cytokines, even in response to 1B3-transfected tumor cells directly. Live cell imaging demonstrated PBMC-mediated cytotoxicity against 1B3-transfected tumor cells. These data demonstrate for the first time that miR-193a-3p induces long-term immunity against tumor development via modulation of the tumor microenvironment and induction of immunogenic cell death.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"470-485"},"PeriodicalIF":0.0,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247534/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141616906","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-12DOI: 10.18632/oncotarget.28593
Gaurav S Choudhary, Trinh T Tat, Saurav Misra, Brian T Hill, Mitchell R Smith, Alexandru Almasan, Suparna Mazumder
{"title":"Correction: Cyclin E/Cdk2-dependent phosphorylation of Mcl-1 determines its stability and cellular sensitivity to BH3 mimetics.","authors":"Gaurav S Choudhary, Trinh T Tat, Saurav Misra, Brian T Hill, Mitchell R Smith, Alexandru Almasan, Suparna Mazumder","doi":"10.18632/oncotarget.28593","DOIUrl":"10.18632/oncotarget.28593","url":null,"abstract":"","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"468-469"},"PeriodicalIF":0.0,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247532/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141616904","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-10DOI: 10.18632/oncotarget.28607
Maria Gracia-Hernandez, Manasa Suresh, Alejandro Villagra
{"title":"The advances in targeting CD47/SIRPα \"do not eat me\" axis and their ongoing challenges as an anticancer therapy.","authors":"Maria Gracia-Hernandez, Manasa Suresh, Alejandro Villagra","doi":"10.18632/oncotarget.28607","DOIUrl":"10.18632/oncotarget.28607","url":null,"abstract":"","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"462-465"},"PeriodicalIF":0.0,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11235132/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141563995","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-10DOI: 10.18632/oncotarget.28605
Alexandra Jackovich, Barbara J Gitlitz, Justin Wayne Wong Tiu-Lim, Vinay Duddalwar, Kevin George King, Anthony B El-Khoueiry, Jacob Stephen Thomas, Denice Tsao-Wei, David I Quinn, Parkash S Gill, Jorge J Nieva
Objective: Patients with relapsed or metastatic head and neck squamous cell carcinoma (HNSCC) after primary local therapy have low response rates with cetuximab, systemic chemotherapy or check point inhibitor therapy. Novel combination therapies with the potential to improve outcomes for patients with HNSCC is an area of high unmet need.
Methods: This is a phase II single-arm clinical trial of locally advanced or metastatic HNSCC patients treated with a combination of soluble EphB4-human serum albumin (sEphB4-HSA) fusion protein and pembrolizumab after platinum-based chemotherapy with up to 2 prior lines of treatment. The primary endpoints were safety and tolerability and the primary efficacy endpoint was overall response rate (ORR). Secondary endpoints included progression free survival (PFS) and overall survival (OS). HPV status and EphrinB2 expression were evaluated for outcome.
Results: Twenty-five patients were enrolled. Median follow up was 40.4 months (range 9.8 - 40.4). There were 6 responders (ORR 24%). There were 5 responders in the 11 HPV-negative and EphrinB2 positive patients, (ORR 45%) with 2 of these patients achieving a complete response (CR). The median PFS in HPV-negative/EphrinB2 positive patients was 3.2 months (95% CI 1.1, 7.3). Median OS in HPV-negative/EphrinB2 positive patients was 10.9 months (95% CI 2.0, 13.7). Hypertension, transaminitis and fatigue were the most common toxicities.
Discussion: The combination of sEphB4-HSA and pembrolizumab has a favorable toxicity profile and favorable activity particularly among HPV-negative EphrinB2 positive patients with HNSCC.
{"title":"Improved efficacy of pembrolizumab combined with soluble EphB4-albumin in HPV-negative EphrinB2 positive head neck squamous cell carcinoma.","authors":"Alexandra Jackovich, Barbara J Gitlitz, Justin Wayne Wong Tiu-Lim, Vinay Duddalwar, Kevin George King, Anthony B El-Khoueiry, Jacob Stephen Thomas, Denice Tsao-Wei, David I Quinn, Parkash S Gill, Jorge J Nieva","doi":"10.18632/oncotarget.28605","DOIUrl":"10.18632/oncotarget.28605","url":null,"abstract":"<p><strong>Objective: </strong>Patients with relapsed or metastatic head and neck squamous cell carcinoma (HNSCC) after primary local therapy have low response rates with cetuximab, systemic chemotherapy or check point inhibitor therapy. Novel combination therapies with the potential to improve outcomes for patients with HNSCC is an area of high unmet need.</p><p><strong>Methods: </strong>This is a phase II single-arm clinical trial of locally advanced or metastatic HNSCC patients treated with a combination of soluble EphB4-human serum albumin (sEphB4-HSA) fusion protein and pembrolizumab after platinum-based chemotherapy with up to 2 prior lines of treatment. The primary endpoints were safety and tolerability and the primary efficacy endpoint was overall response rate (ORR). Secondary endpoints included progression free survival (PFS) and overall survival (OS). HPV status and EphrinB2 expression were evaluated for outcome.</p><p><strong>Results: </strong>Twenty-five patients were enrolled. Median follow up was 40.4 months (range 9.8 - 40.4). There were 6 responders (ORR 24%). There were 5 responders in the 11 HPV-negative and EphrinB2 positive patients, (ORR 45%) with 2 of these patients achieving a complete response (CR). The median PFS in HPV-negative/EphrinB2 positive patients was 3.2 months (95% CI 1.1, 7.3). Median OS in HPV-negative/EphrinB2 positive patients was 10.9 months (95% CI 2.0, 13.7). Hypertension, transaminitis and fatigue were the most common toxicities.</p><p><strong>Discussion: </strong>The combination of sEphB4-HSA and pembrolizumab has a favorable toxicity profile and favorable activity particularly among HPV-negative EphrinB2 positive patients with HNSCC.</p>","PeriodicalId":19499,"journal":{"name":"Oncotarget","volume":"15 ","pages":"444-458"},"PeriodicalIF":0.0,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11235133/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141563993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}