首页 > 最新文献

Clinical Cancer Research最新文献

英文 中文
Patient-derived Organoids and Xenografts Uncover Therapeutic Vulnerabilities in Colorectal Signet Ring Cell Carcinomas
IF 11.5 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-29 DOI: 10.1158/1078-0432.ccr-24-2329
Nazia Chaudhary, Alessandro La Ferlita, Bhagya Shree. Choudhary, Eeshrita Jog, Mufaddal Kazi, Showket Yahya, Afiya Dalwai, Vikas Ostwal, Satishkumar Kumar, Siddhi Redkar, Nileema Khapare, Vaishali Kailaje, Akshaya B, Poonam Gera, Munita Bal, Nandini Verma, Rahul Thorat, Avanish Saklani, Lalit Sehgal, Sorab N. Dalal
Purpose: Identifying therapeutic targets for Signet Ring Cell Carcinoma (SRCC) of the colon and rectum is a clinical challenge due to the lack of Patient-Derived Organoids (PDO) or Xenografts (PDX). We present a robust method to establish PDO and PDX models to answer address this unmet need. We demonstrate that these models identify novel therapeutic strategies targeting therapy resistance and peritoneal metastasis. Experimental Design: We derived nine PDO and PDX models from colorectal SRCC patients. Detailed histopathological characterization confirmed the fidelity of these models to the original tumors. Drug sensitivity assays were conducted in-vitro and in-vivo to assess therapeutic efficacy and impact on peritoneal metastasis. An RNA-seq analysis was performed to identify critical pathways contributing to therapy resistance and metastatic progression. Results: We successfully developed and characterized PDO and PDX models from nine SRCC patients. The SRCC PDO and PDX models exhibited histopathological features consistent with the original tumors, including high mucin content and eccentric nuclei. They demonstrated increased sensitivity to FOLFIRI combined with Paclitaxel or vincristine, reducing peritoneal metastasis. RNA-seq analysis revealed the upregulation of autophagy genes in SRCC. Treatment with Chloroquine alone resulted in decreased tumor growth and peritoneal metastasis. Conclusions: Our study establishes PDO and PDX models as robust platforms for studying SRCC and identifying potential therapeutic strategies. Combining FOLFIRI with paclitaxel/ vincristine or Chloroquine alone inhibits tumor growth and prevents peritoneal metastasis, showing promise for clinical translation. These findings suggest that combining FOLFIRI with IP paclitaxel warrants further investigation in Phase-I clinical trials for SRCC patients.
{"title":"Patient-derived Organoids and Xenografts Uncover Therapeutic Vulnerabilities in Colorectal Signet Ring Cell Carcinomas","authors":"Nazia Chaudhary, Alessandro La Ferlita, Bhagya Shree. Choudhary, Eeshrita Jog, Mufaddal Kazi, Showket Yahya, Afiya Dalwai, Vikas Ostwal, Satishkumar Kumar, Siddhi Redkar, Nileema Khapare, Vaishali Kailaje, Akshaya B, Poonam Gera, Munita Bal, Nandini Verma, Rahul Thorat, Avanish Saklani, Lalit Sehgal, Sorab N. Dalal","doi":"10.1158/1078-0432.ccr-24-2329","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-24-2329","url":null,"abstract":"Purpose: Identifying therapeutic targets for Signet Ring Cell Carcinoma (SRCC) of the colon and rectum is a clinical challenge due to the lack of Patient-Derived Organoids (PDO) or Xenografts (PDX). We present a robust method to establish PDO and PDX models to answer address this unmet need. We demonstrate that these models identify novel therapeutic strategies targeting therapy resistance and peritoneal metastasis. Experimental Design: We derived nine PDO and PDX models from colorectal SRCC patients. Detailed histopathological characterization confirmed the fidelity of these models to the original tumors. Drug sensitivity assays were conducted in-vitro and in-vivo to assess therapeutic efficacy and impact on peritoneal metastasis. An RNA-seq analysis was performed to identify critical pathways contributing to therapy resistance and metastatic progression. Results: We successfully developed and characterized PDO and PDX models from nine SRCC patients. The SRCC PDO and PDX models exhibited histopathological features consistent with the original tumors, including high mucin content and eccentric nuclei. They demonstrated increased sensitivity to FOLFIRI combined with Paclitaxel or vincristine, reducing peritoneal metastasis. RNA-seq analysis revealed the upregulation of autophagy genes in SRCC. Treatment with Chloroquine alone resulted in decreased tumor growth and peritoneal metastasis. Conclusions: Our study establishes PDO and PDX models as robust platforms for studying SRCC and identifying potential therapeutic strategies. Combining FOLFIRI with paclitaxel/ vincristine or Chloroquine alone inhibits tumor growth and prevents peritoneal metastasis, showing promise for clinical translation. These findings suggest that combining FOLFIRI with IP paclitaxel warrants further investigation in Phase-I clinical trials for SRCC patients.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"476 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143056490","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}
引用次数: 0
Targeting AXL inhibits the growth and metastasis of prostate cancer in bone
IF 11.5 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-29 DOI: 10.1158/1078-0432.ccr-24-3028
Chun-Lung Chiu, Dalin Zhang, Hongjuan Zhao, Yi Wei, Alexandra Lapat. Polasko, Mikkel Thy. Thomsen, Vanessa Yang, Kasie Kexin. Yang, Spencer Hauck, Eric E. Peterson, Ru M. Wen, Zhengyuan Qiu, Eva Corey, Yu Rebecca Miao, Erinn B. Rankin, Donna M. Peehl, Jiaoti Huang, Amato J. Giaccia, James D. Brooks
Purpose: After failing primary and secondary hormonal therapy, castration-resistant and neuroendocrine prostate cancer metastatic to the bone is invariably lethal, although treatment with docetaxel and carboplatin can modestly improve survival. Therefore, agents targeting biologically relevant pathways in PCa and potentially synergizing with docetaxel and carboplatin in inhibiting bone metastasis growth are urgently needed. Experimental Design: Phosphorylated (activated) AXL expression in human prostate cancer bone metastases was assessed by immunohistochemical staining. We evaluated the effects of a novel soluble AXL signaling inhibitor, sAXL (batiraxcept or AVB-S6-500), on the tumor growth and lung metastases in PCa patient-derived xenograft models (PDX) that implanted intratibally. After injection of LuCaP cells into the tibiae, tumors were treated with batiraxcept and docetaxel or carboplatin alone or in combination, and tumor growth was monitored by serum PSA or bioluminescence. Tumor burden was quantified by human-specific Ku70 staining, and metastasis to the lung was determined using qPCR. Transcriptomic profiling, western blotting and immunohistochemistry were performed to identify treatment-regulated gene and protein profile changes. Results: High AXL phosphorylation in human PCa bone metastases correlated with shortened survival. Batiraxcept alone or in combination with docetaxel or carboplatin significantly suppressed intratibial tumor growth and suppressed metastasis to the lung through multiple mechanisms, including repression of cancer stemness genes (CD44, ALDH1A1, TACSTD2, ATXN1) and the PI3K, JAK, MAPK, and E2F1/NUSAP1 signaling pathways. Conclusions: Our study provides a robust preclinical rationale and mechanisms of action for using batiraxcept as a single agent or in combination with docetaxel or carboplatin to treat lethal mPCa.
{"title":"Targeting AXL inhibits the growth and metastasis of prostate cancer in bone","authors":"Chun-Lung Chiu, Dalin Zhang, Hongjuan Zhao, Yi Wei, Alexandra Lapat. Polasko, Mikkel Thy. Thomsen, Vanessa Yang, Kasie Kexin. Yang, Spencer Hauck, Eric E. Peterson, Ru M. Wen, Zhengyuan Qiu, Eva Corey, Yu Rebecca Miao, Erinn B. Rankin, Donna M. Peehl, Jiaoti Huang, Amato J. Giaccia, James D. Brooks","doi":"10.1158/1078-0432.ccr-24-3028","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-24-3028","url":null,"abstract":"Purpose: After failing primary and secondary hormonal therapy, castration-resistant and neuroendocrine prostate cancer metastatic to the bone is invariably lethal, although treatment with docetaxel and carboplatin can modestly improve survival. Therefore, agents targeting biologically relevant pathways in PCa and potentially synergizing with docetaxel and carboplatin in inhibiting bone metastasis growth are urgently needed. Experimental Design: Phosphorylated (activated) AXL expression in human prostate cancer bone metastases was assessed by immunohistochemical staining. We evaluated the effects of a novel soluble AXL signaling inhibitor, sAXL (batiraxcept or AVB-S6-500), on the tumor growth and lung metastases in PCa patient-derived xenograft models (PDX) that implanted intratibally. After injection of LuCaP cells into the tibiae, tumors were treated with batiraxcept and docetaxel or carboplatin alone or in combination, and tumor growth was monitored by serum PSA or bioluminescence. Tumor burden was quantified by human-specific Ku70 staining, and metastasis to the lung was determined using qPCR. Transcriptomic profiling, western blotting and immunohistochemistry were performed to identify treatment-regulated gene and protein profile changes. Results: High AXL phosphorylation in human PCa bone metastases correlated with shortened survival. Batiraxcept alone or in combination with docetaxel or carboplatin significantly suppressed intratibial tumor growth and suppressed metastasis to the lung through multiple mechanisms, including repression of cancer stemness genes (CD44, ALDH1A1, TACSTD2, ATXN1) and the PI3K, JAK, MAPK, and E2F1/NUSAP1 signaling pathways. Conclusions: Our study provides a robust preclinical rationale and mechanisms of action for using batiraxcept as a single agent or in combination with docetaxel or carboplatin to treat lethal mPCa.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"54 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143056489","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}
引用次数: 0
LINE-1 Retrotransposons as Cell-free DNA Biomarkers for Multi-Cancer Early Detection
IF 11.5 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-29 DOI: 10.1158/1078-0432.ccr-24-4051
Jeffrey J. Szymanski, Pradeep S. Chauhan, Aadel A. Chaudhuri
LINE-1 retrotransposons, comprising 17% of the genome, drive cancer instability through hypomethylation. The DIAMOND assay, targeting LINE-1 hypomethylation with bisulfite sequencing of cell free DNA, achieved AUCs of 88% to 100% across six cancer types, surpassing mutation-based diagnostics and suggesting utility in early cancer detection and management.
{"title":"LINE-1 Retrotransposons as Cell-free DNA Biomarkers for Multi-Cancer Early Detection","authors":"Jeffrey J. Szymanski, Pradeep S. Chauhan, Aadel A. Chaudhuri","doi":"10.1158/1078-0432.ccr-24-4051","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-24-4051","url":null,"abstract":"LINE-1 retrotransposons, comprising 17% of the genome, drive cancer instability through hypomethylation. The DIAMOND assay, targeting LINE-1 hypomethylation with bisulfite sequencing of cell free DNA, achieved AUCs of 88% to 100% across six cancer types, surpassing mutation-based diagnostics and suggesting utility in early cancer detection and management.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"10 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143056180","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}
引用次数: 0
Time dependency for human papillomavirus circulating tumor DNA detection after chemoradiation as a prognostic biomarker for localized anal cancer. 化疗后人类乳头瘤病毒循环肿瘤 DNA 检测作为局部肛门癌预后生物标志物的时间依赖性。
IF 1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-28 DOI: 10.1158/1078-0432.CCR-24-2575
Van K Morris, Weihong Xiao, Kangyu Lin, Chi Wut Wong, Michael T Wotman, Emma B Holliday, Ryan W Huey, Sonal S Noticewala, Ethan B Ludmir, Alisha H Bent, Kaysia Ludford, Craig Messick, Eugene J Koay, Grace Smith, Tsuyoshi Konishi, Brian Bednarski, George J Chang, Albert C Koong, Y Nancy You, Prajnan Das, Maura L Gillison

Background: While detection of circulating tumor DNA (ctDNA) weeks after surgery is linked to recurrence for other solid tumors, the optimal time point for ctDNA assessment as a prognostic biomarker following chemoradiation for anal cancer is undefined.

Methods: Patients with stages I-III anal cancer treated with chemoradiation between 12/2020-5/2024 were evaluated for HPV ctDNA status at baseline, at the end of chemoradiation, and during surveillance using a droplet digital HPV ctDNA PCR assay targeting HPV E6 and E7 oncogenes for 13 oncogenic HPV types. Median recurrence-free survival (RFS) according to HPV ctDNA status was estimated via Kaplan-Meier and compared using a log-rank test.

Results: Detection of HPV ctDNA at ≥3 months after chemoradiation was associated with recurrence (80% versus 2%; odds ratio 168, 95% CI 13.6-2080; p<.0001) and inferior RFS (4.9 months versus not reached (NR); hazard ratio (HR) 39.2, 95% CI 4.6-330; p<.0001) relative to HPV ctDNA-negative status. Sensitivity and specificity for recurrence according to HPV ctDNA detection were 89% and 95%, respectively, with positive and negative predictive values of 80% and 98%, respectively. Differences in RFS according to HPV ctDNA status were not observed at the end of treatment (median RFS NR for both; HR 1.6, 95% CI .35-7.4; p=.48).

Conclusions: With a novel, highly sensitive assay, detection of HPV ctDNA at least 3 months after chemoradiation was associated with unfavorable survival. Future clinical trials should incorporate this 3-month post-treatment time point to identify patients with HPV-positive anal cancer at elevated recurrence risk according to HPV ctDNA status.

背景:虽然术后数周检测循环肿瘤DNA(ctDNA)与其他实体瘤的复发有关,但ctDNA评估作为肛门癌化疗后预后生物标志物的最佳时间点尚未确定:在2020年12月至2024年5月期间接受化疗的I-III期肛门癌患者在基线、化疗结束时和监测期间使用针对13种致癌HPV类型的HPV E6和E7致癌基因的液滴数字HPV ctDNA PCR检测法评估HPV ctDNA状态。根据HPV ctDNA状态的中位无复发生存期(RFS)通过Kaplan-Meier进行估算,并使用对数秩检验进行比较:结果:化疗后≥3个月时检测到HPV ctDNA与复发有关(80%对2%;几率比168,95% CI 13.6-2080;p结论:通过一种新型、高灵敏度的检测方法,在化疗后至少 3 个月检测到 HPV ctDNA 与不利的生存率相关。未来的临床试验应结合治疗后 3 个月的时间点,根据 HPV ctDNA 状态确定复发风险较高的 HPV 阳性肛门癌患者。
{"title":"Time dependency for human papillomavirus circulating tumor DNA detection after chemoradiation as a prognostic biomarker for localized anal cancer.","authors":"Van K Morris, Weihong Xiao, Kangyu Lin, Chi Wut Wong, Michael T Wotman, Emma B Holliday, Ryan W Huey, Sonal S Noticewala, Ethan B Ludmir, Alisha H Bent, Kaysia Ludford, Craig Messick, Eugene J Koay, Grace Smith, Tsuyoshi Konishi, Brian Bednarski, George J Chang, Albert C Koong, Y Nancy You, Prajnan Das, Maura L Gillison","doi":"10.1158/1078-0432.CCR-24-2575","DOIUrl":"https://doi.org/10.1158/1078-0432.CCR-24-2575","url":null,"abstract":"<p><strong>Background: </strong>While detection of circulating tumor DNA (ctDNA) weeks after surgery is linked to recurrence for other solid tumors, the optimal time point for ctDNA assessment as a prognostic biomarker following chemoradiation for anal cancer is undefined.</p><p><strong>Methods: </strong>Patients with stages I-III anal cancer treated with chemoradiation between 12/2020-5/2024 were evaluated for HPV ctDNA status at baseline, at the end of chemoradiation, and during surveillance using a droplet digital HPV ctDNA PCR assay targeting HPV E6 and E7 oncogenes for 13 oncogenic HPV types. Median recurrence-free survival (RFS) according to HPV ctDNA status was estimated via Kaplan-Meier and compared using a log-rank test.</p><p><strong>Results: </strong>Detection of HPV ctDNA at ≥3 months after chemoradiation was associated with recurrence (80% versus 2%; odds ratio 168, 95% CI 13.6-2080; p<.0001) and inferior RFS (4.9 months versus not reached (NR); hazard ratio (HR) 39.2, 95% CI 4.6-330; p<.0001) relative to HPV ctDNA-negative status. Sensitivity and specificity for recurrence according to HPV ctDNA detection were 89% and 95%, respectively, with positive and negative predictive values of 80% and 98%, respectively. Differences in RFS according to HPV ctDNA status were not observed at the end of treatment (median RFS NR for both; HR 1.6, 95% CI .35-7.4; p=.48).</p><p><strong>Conclusions: </strong>With a novel, highly sensitive assay, detection of HPV ctDNA at least 3 months after chemoradiation was associated with unfavorable survival. Future clinical trials should incorporate this 3-month post-treatment time point to identify patients with HPV-positive anal cancer at elevated recurrence risk according to HPV ctDNA status.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":10.0,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143051845","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}
引用次数: 0
Receptor CDCP1 is a potential target for personalized imaging and treatment of poor outcome HER2+, triple negative and metastatic ER+/HER2- breast cancers.
IF 1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-27 DOI: 10.1158/1078-0432.CCR-24-2865
Madeline Gough, Kayden Kx Kwah, Tashbib Khan, Saikat Ghosh, Biao Sun, Catherine Yj Lee, Kamil A Sokolowski, Brian Wc Tse, Lashith Wickramasuriya, Kaltin Ferguson, Rebecca Rogers, Justin B Goh, Nicholas L Fletcher, Zachary H Houston, Kristofer J Thurecht, Laura J Bray, Cheng Liu, Christopher Pyke, Elgene Lim, Cameron E Snell, Yaowu He, John D Hooper, Thomas Kryza

Purpose: Receptor CUB-domain containing- protein 1 (CDCP1) was evaluated as a target for detection and treatment of breast cancer.

Experimental design: CDCP1 expression was assessed immunohistochemically in tumors from 423 patients (119 triple-negative breast cancer (TNBC); 75 HER2+; 229 ER+/HER2- including 228 primary tumors, 229 lymph node and 47 distant metastases). Cell cytotoxicity induced in vitro by a CDCP1-targeting antibody-drug conjugate (ADC), consisting of the human/mouse chimeric antibody ch10D7 and the microtubule disruptor monomethyl auristatin E (MMAE), was quantified, including in combination with HER2-targeting ADC T-DM1. Detection of CDCP1-expressing primary and metastatic xenografts in mice was examined by PET-CT imaging using 89zirconium-labelled ch10D7 (89Zr-ch10D7). The impact of ch10D7-MMAE on tumor burden and survival in vivo, including in combination with T-DM1, was quantified in cell line and patient-derived xenograft mouse models.

Results: CDCP1 is expressed predominantly on the surface of malignant cells of 70% of TNBCs, 80% of HER2+ tumors, and increases in ER+/HER2- tumors from 44.9% in primary tumors to 56.4% in lymph node metastases and 74.3% in distant metastases. PET-CT imaging with 89Zr-ch10D7 is effective for the detection of primary and metastatic CDCP1-expressing TNBCs in mice. ADC ch10D7-MMAE kills CDCP1-expressing cells in vitro and controls primary and metastatic TNBC xenografts in mice, conferring significant survival advantages over chemotherapy. It compares favorably to T-DM1 in vivo, and ch10D7-MMAE combined with T-DM1 showed the most potent efficacy, markedly reducing tumor burden of CDCP1+/HER2+ xenografts and prolonging mouse survival, compared with T-DM1 or ch10D7.

Conclusions: CDCP1-directed molecular imaging has potential to identify aggressive breast cancers for CDCP1-targeted treatment.

{"title":"Receptor CDCP1 is a potential target for personalized imaging and treatment of poor outcome HER2+, triple negative and metastatic ER+/HER2- breast cancers.","authors":"Madeline Gough, Kayden Kx Kwah, Tashbib Khan, Saikat Ghosh, Biao Sun, Catherine Yj Lee, Kamil A Sokolowski, Brian Wc Tse, Lashith Wickramasuriya, Kaltin Ferguson, Rebecca Rogers, Justin B Goh, Nicholas L Fletcher, Zachary H Houston, Kristofer J Thurecht, Laura J Bray, Cheng Liu, Christopher Pyke, Elgene Lim, Cameron E Snell, Yaowu He, John D Hooper, Thomas Kryza","doi":"10.1158/1078-0432.CCR-24-2865","DOIUrl":"https://doi.org/10.1158/1078-0432.CCR-24-2865","url":null,"abstract":"<p><strong>Purpose: </strong>Receptor CUB-domain containing- protein 1 (CDCP1) was evaluated as a target for detection and treatment of breast cancer.</p><p><strong>Experimental design: </strong>CDCP1 expression was assessed immunohistochemically in tumors from 423 patients (119 triple-negative breast cancer (TNBC); 75 HER2+; 229 ER+/HER2- including 228 primary tumors, 229 lymph node and 47 distant metastases). Cell cytotoxicity induced in vitro by a CDCP1-targeting antibody-drug conjugate (ADC), consisting of the human/mouse chimeric antibody ch10D7 and the microtubule disruptor monomethyl auristatin E (MMAE), was quantified, including in combination with HER2-targeting ADC T-DM1. Detection of CDCP1-expressing primary and metastatic xenografts in mice was examined by PET-CT imaging using 89zirconium-labelled ch10D7 (89Zr-ch10D7). The impact of ch10D7-MMAE on tumor burden and survival in vivo, including in combination with T-DM1, was quantified in cell line and patient-derived xenograft mouse models.</p><p><strong>Results: </strong>CDCP1 is expressed predominantly on the surface of malignant cells of 70% of TNBCs, 80% of HER2+ tumors, and increases in ER+/HER2- tumors from 44.9% in primary tumors to 56.4% in lymph node metastases and 74.3% in distant metastases. PET-CT imaging with 89Zr-ch10D7 is effective for the detection of primary and metastatic CDCP1-expressing TNBCs in mice. ADC ch10D7-MMAE kills CDCP1-expressing cells in vitro and controls primary and metastatic TNBC xenografts in mice, conferring significant survival advantages over chemotherapy. It compares favorably to T-DM1 in vivo, and ch10D7-MMAE combined with T-DM1 showed the most potent efficacy, markedly reducing tumor burden of CDCP1+/HER2+ xenografts and prolonging mouse survival, compared with T-DM1 or ch10D7.</p><p><strong>Conclusions: </strong>CDCP1-directed molecular imaging has potential to identify aggressive breast cancers for CDCP1-targeted treatment.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":10.0,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045557","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}
引用次数: 0
Impact of Race/Ethnicity on Clinical and Genomic Characteristics, Trial Participation, and Genotype-Matched Therapy among Patients with Metastatic Breast Cancer.
IF 1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-27 DOI: 10.1158/1078-0432.CCR-24-2825
Rupali Sood, Andrzej Niemierko, Lianne Ryan, Laura Spring, Beverly Moy, Aditya Bardia, Neelima Vidula

Background: Race/ethnicity may affect outcomes in metastatic breast cancer (MBC) due to biological and social determinants. We evaluated the impact of race/ethnicity on clinical, socioeconomic, and genomic characteristics, clinical trial participation, and receipt of genotype-matched therapy among patients with MBC.

Methods: A retrospective study of patients with MBC who underwent cell-free DNA testing (cfDNA, Guardant360â, 74 gene panel) between 11/2016 and 11/2020 was conducted. Receipt of genotype-matched therapy targeted at a cfDNA actionable mutation was determined. Pearson's chi-squared and Wilcoxon rank-sum tests were used to compare categorical and continuous variables between groups. Multivariable logistic regression was used to assess the association of race and receiving matched therapy.

Results: 425 patients with MBC and cfDNA results were identified (White: 369, Black: 27, Hispanic 15, and Asian 14). White patients traveled further for cancer care than other groups (p<0.001). White patients had the highest rates of commercial insurance, Black patients had the highest rates of state-supported insurance, and Asian patients had the highest uninsured rates (p<0.001). Clinical trial enrollment did not differ by race/ethnicity (p=0.34). The proportion of patients with ≥1 actionable mutation in cfDNA did not vary by race/ethnicity (p=0.18). The highest rates of matched therapy were observed in White patients (p<0.001). After multivariable logistic regression adjusting for subtype, commercial vs. other insurance, Charlson comorbidity index, and distance to center, White patients remained more likely to receive matched therapy (p=0.024).

Conclusions: Racial/ethnic minority patients were less likely to receive matched therapy. Further research is needed to identify barriers to precision medicine.

{"title":"Impact of Race/Ethnicity on Clinical and Genomic Characteristics, Trial Participation, and Genotype-Matched Therapy among Patients with Metastatic Breast Cancer.","authors":"Rupali Sood, Andrzej Niemierko, Lianne Ryan, Laura Spring, Beverly Moy, Aditya Bardia, Neelima Vidula","doi":"10.1158/1078-0432.CCR-24-2825","DOIUrl":"https://doi.org/10.1158/1078-0432.CCR-24-2825","url":null,"abstract":"<p><strong>Background: </strong>Race/ethnicity may affect outcomes in metastatic breast cancer (MBC) due to biological and social determinants. We evaluated the impact of race/ethnicity on clinical, socioeconomic, and genomic characteristics, clinical trial participation, and receipt of genotype-matched therapy among patients with MBC.</p><p><strong>Methods: </strong>A retrospective study of patients with MBC who underwent cell-free DNA testing (cfDNA, Guardant360â, 74 gene panel) between 11/2016 and 11/2020 was conducted. Receipt of genotype-matched therapy targeted at a cfDNA actionable mutation was determined. Pearson's chi-squared and Wilcoxon rank-sum tests were used to compare categorical and continuous variables between groups. Multivariable logistic regression was used to assess the association of race and receiving matched therapy.</p><p><strong>Results: </strong>425 patients with MBC and cfDNA results were identified (White: 369, Black: 27, Hispanic 15, and Asian 14). White patients traveled further for cancer care than other groups (p<0.001). White patients had the highest rates of commercial insurance, Black patients had the highest rates of state-supported insurance, and Asian patients had the highest uninsured rates (p<0.001). Clinical trial enrollment did not differ by race/ethnicity (p=0.34). The proportion of patients with ≥1 actionable mutation in cfDNA did not vary by race/ethnicity (p=0.18). The highest rates of matched therapy were observed in White patients (p<0.001). After multivariable logistic regression adjusting for subtype, commercial vs. other insurance, Charlson comorbidity index, and distance to center, White patients remained more likely to receive matched therapy (p=0.024).</p><p><strong>Conclusions: </strong>Racial/ethnic minority patients were less likely to receive matched therapy. Further research is needed to identify barriers to precision medicine.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":10.0,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143045551","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}
引用次数: 0
Follow-Up Analysis Enhances Understanding of Molecular Residual Disease in Localized Non-Small Cell Lung Cancer
IF 11.5 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-24 DOI: 10.1158/1078-0432.ccr-24-2909
Jia-Tao Zhang, Si-Yang Liu, Xuan Gao, Si-Yang Maggie. Liu, Bingfa Yan, Chen Huang, Zicong Jiao, Hong-Hong Yan, Yi Pan, Song Dong, Wei Gao, Yuhua Gong, Hai-Yan Tu, Xue-Feng Xia, Qing Zhou, Wen-Zhao Zhong, Xue-Ning Yang, Xin Yi, Yi-Long Wu
Purpose: The prognostic value of molecular residual disease (MRD) in non-small cell lung cancer (NSCLC) is well-established, with treatment-guiding results anticipated. Here, we present updated analyses from our previous published cohort study of 261 NSCLC patients undergoing complete resection. Experimental Design: 261 patients with stage I-III lung cancer who underwent radical surgery were enrolled. Enrolled patients underwent follow-up blood draws according to the predefined time points after surgery. As of December 31st, 2023, with a median follow-up of 43.4 months, 948 postoperative blood samples were collected. Results: Landmark and longitudinal MRD exhibited positive predictive values of 91.3% and 92.8%, respectively, with a median lead times of 5.2 months. Negative predictive values were 76.5% and 93.2%, respectively. Patients with landmark undetectable MRD could not be benefited from adjuvant therapy through the updated follow-up (p=0.529). Among the 13 patients with recurrent NSCLC and longitudinal undetectable MRD, seven (53.8%) had brain-only metastases, and four (30.8%) had no updated blood samples for over six months prior to recurrence. Besides, for those with longitudinal detectable MRD, higher maximum variant allele frequency (&gt;0.55%) and ctDNA level (&gt;13 hGE/ml) were associated with a high risk of short-term recurrence. Additionally, updated follow-up data further support the peak time for detectable MRD was 18 months after landmark detection. Conclusions: These findings suggest the significant potential of MRD in guiding personalized treatment for NSCLC. And postoperative longitudinal undetectable MRD can indicate a cured population.
{"title":"Follow-Up Analysis Enhances Understanding of Molecular Residual Disease in Localized Non-Small Cell Lung Cancer","authors":"Jia-Tao Zhang, Si-Yang Liu, Xuan Gao, Si-Yang Maggie. Liu, Bingfa Yan, Chen Huang, Zicong Jiao, Hong-Hong Yan, Yi Pan, Song Dong, Wei Gao, Yuhua Gong, Hai-Yan Tu, Xue-Feng Xia, Qing Zhou, Wen-Zhao Zhong, Xue-Ning Yang, Xin Yi, Yi-Long Wu","doi":"10.1158/1078-0432.ccr-24-2909","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-24-2909","url":null,"abstract":"Purpose: The prognostic value of molecular residual disease (MRD) in non-small cell lung cancer (NSCLC) is well-established, with treatment-guiding results anticipated. Here, we present updated analyses from our previous published cohort study of 261 NSCLC patients undergoing complete resection. Experimental Design: 261 patients with stage I-III lung cancer who underwent radical surgery were enrolled. Enrolled patients underwent follow-up blood draws according to the predefined time points after surgery. As of December 31st, 2023, with a median follow-up of 43.4 months, 948 postoperative blood samples were collected. Results: Landmark and longitudinal MRD exhibited positive predictive values of 91.3% and 92.8%, respectively, with a median lead times of 5.2 months. Negative predictive values were 76.5% and 93.2%, respectively. Patients with landmark undetectable MRD could not be benefited from adjuvant therapy through the updated follow-up (p=0.529). Among the 13 patients with recurrent NSCLC and longitudinal undetectable MRD, seven (53.8%) had brain-only metastases, and four (30.8%) had no updated blood samples for over six months prior to recurrence. Besides, for those with longitudinal detectable MRD, higher maximum variant allele frequency (&amp;gt;0.55%) and ctDNA level (&amp;gt;13 hGE/ml) were associated with a high risk of short-term recurrence. Additionally, updated follow-up data further support the peak time for detectable MRD was 18 months after landmark detection. Conclusions: These findings suggest the significant potential of MRD in guiding personalized treatment for NSCLC. And postoperative longitudinal undetectable MRD can indicate a cured population.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"58 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143030988","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}
引用次数: 0
Vvax001, a Therapeutic Vaccine for Patients with HPV16-positive High-grade Cervical Intraepithelial Neoplasia: a Phase II Trial
IF 11.5 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-24 DOI: 10.1158/1078-0432.ccr-24-1662
Anneke L. Eerkens, Martha D. Esajas, Koen Brummel, Annegé Vledder, Nienke van Rooij, Annechien Plat, Stefany B. Avalos Haro, Sterre T. Paijens, Lorian Slagter-Menkema, Ed Schuuring, Naomi Werner, Jos G.W. Kosterink, Bart-Jan Kroesen, Jan C. Wilschut, Toos Daemen, Joost Bart, Hans W. Nijman, Marco de Bruyn, Refika Yigit
Purpose: Human papillomavirus (HPV) infection is the major cause of (pre)malignant cervical lesions. We previously demonstrated that Vvax001, a replication-incompetent Semliki Forest virus (SFV) vaccine encoding HPV type 16 (HPV16) E6 and E7, induced potent anti-E6 and -E7 cytotoxic T-cell responses. Here, we investigated the clinical efficacy of Vvax001 in patients with HPV16-positive cervical intraepithelial neoplasia grade 3 (CIN3). Patients and Methods: Patients with newly diagnosed HPV16-positive CIN3 were eligible for participation. Patients received 3 immunizations of Vvax001 (5x107 infectious particles) at a three-week interval. Up to 19 weeks after the last immunization patients were monitored for regression of CIN3 by colposcopy. A colposcopy-guided biopsy was taken at the last visit and a standard of care loop excision was performed only in case of remaining CIN2/3. Histopathologic response rates, HPV16 clearance, treatment-related adverse events (trAEs), and vaccine-induced immune responses were assessed. Results: A total of 18 patients were enrolled and fully immunized. Colposcopic examination revealed a reduction in CIN3 lesion sizes in 17/18 patients (94%) already evident from 3 weeks onwards after the last immunization. A histopathological complete response (regression to CIN1 or no dysplasia) was observed in 9/18 patients (50%), and HPV16 clearance in 10/16 patients (63%). Vvax001 did not induce clearance of other HPV types. To date, no recurrences have been observed, with a median and longest disease-free survival of 20 and 30 months, respectively. No serious AEs were observed. Conclusions: Treatment with Vvax001 is safe, feasible, and shows preliminary clinical effectiveness in patients with HPV16-associated CIN3 lesions.
{"title":"Vvax001, a Therapeutic Vaccine for Patients with HPV16-positive High-grade Cervical Intraepithelial Neoplasia: a Phase II Trial","authors":"Anneke L. Eerkens, Martha D. Esajas, Koen Brummel, Annegé Vledder, Nienke van Rooij, Annechien Plat, Stefany B. Avalos Haro, Sterre T. Paijens, Lorian Slagter-Menkema, Ed Schuuring, Naomi Werner, Jos G.W. Kosterink, Bart-Jan Kroesen, Jan C. Wilschut, Toos Daemen, Joost Bart, Hans W. Nijman, Marco de Bruyn, Refika Yigit","doi":"10.1158/1078-0432.ccr-24-1662","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-24-1662","url":null,"abstract":"Purpose: Human papillomavirus (HPV) infection is the major cause of (pre)malignant cervical lesions. We previously demonstrated that Vvax001, a replication-incompetent Semliki Forest virus (SFV) vaccine encoding HPV type 16 (HPV16) E6 and E7, induced potent anti-E6 and -E7 cytotoxic T-cell responses. Here, we investigated the clinical efficacy of Vvax001 in patients with HPV16-positive cervical intraepithelial neoplasia grade 3 (CIN3). Patients and Methods: Patients with newly diagnosed HPV16-positive CIN3 were eligible for participation. Patients received 3 immunizations of Vvax001 (5x107 infectious particles) at a three-week interval. Up to 19 weeks after the last immunization patients were monitored for regression of CIN3 by colposcopy. A colposcopy-guided biopsy was taken at the last visit and a standard of care loop excision was performed only in case of remaining CIN2/3. Histopathologic response rates, HPV16 clearance, treatment-related adverse events (trAEs), and vaccine-induced immune responses were assessed. Results: A total of 18 patients were enrolled and fully immunized. Colposcopic examination revealed a reduction in CIN3 lesion sizes in 17/18 patients (94%) already evident from 3 weeks onwards after the last immunization. A histopathological complete response (regression to CIN1 or no dysplasia) was observed in 9/18 patients (50%), and HPV16 clearance in 10/16 patients (63%). Vvax001 did not induce clearance of other HPV types. To date, no recurrences have been observed, with a median and longest disease-free survival of 20 and 30 months, respectively. No serious AEs were observed. Conclusions: Treatment with Vvax001 is safe, feasible, and shows preliminary clinical effectiveness in patients with HPV16-associated CIN3 lesions.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"2 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143027223","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}
引用次数: 0
Intratumoral or Subcutaneous MK-2118, a Non-Cyclic Dinucleotide STING Agonist, With or Without Pembrolizumab for Advanced or Metastatic Solid Tumors or Lymphomas.
IF 1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-23 DOI: 10.1158/1078-0432.CCR-24-2824
Jason J Luke, Randy F Sweis, J Randolph Hecht, Reva Schneider, Mark N Stein, Talia Golan, Timothy A Yap, Anuradha Khilnani, Mo Huang, Runchen Zhao, Thomas Jemielita, Sandip Pravin Patel

Background: We evaluated the non-cyclic dinucleotide stimulator of interferon genes agonist MK-2118 ± pembrolizumab in patients with advanced solid tumors or lymphomas.

Methods: This first-in-human study (NCT03249792) enrolled patients with refractory, advanced solid tumors or lymphomas. Patients received intratumoral (IT) MK-2118 100-20,000 µg (arm 1), IT MK-2118 900-15,000 µg plus intravenous (IV) pembrolizumab 200 mg every 3 weeks (Q3W; arm 2), or subcutaneous (SC) MK-2118 5000-150,000 µg plus IV pembrolizumab 200 mg Q3W (arm 4); arm 3 (visceral injection of MK-2118) was not pursued. IT dosing used an accelerated titration design and modified toxicity probability interval method; SC dosing (arm 4) was started subsequent to arms 1 and 2. Primary objectives were safety/tolerability. MK-2118 pharmacokinetics were a secondary endpoint; objective responses and biomarkers were exploratory endpoints.

Results: 140 patients were enrolled (arm 1, n=27; arm 2, n=57; arm 4, n=56). Grade 3/4 treatment-related adverse events occurred in 22%, 23%, and 11% of patients, respectively, but no maximum tolerated dose was identified up to MK-2118 20,000, 15,000, and 150,000 µg across the 3 arms. Dose-dependent increases in MK-2118 systemic exposure were observed following IT and SC administration. Objective responses were seen in 0%, 6%, and 4% of patients, respectively. IT MK-2118 led to dose-dependent changes in STING-based blood RNA expression levels, interferon-gamma, interferon-gamma‒induced protein 10, and interleukin-6; SC MK-2118 did not generate dose-related immune responses.

Conclusion: IT MK-2118 ± pembrolizumab and SC MK-2118 plus pembrolizumab had manageable toxicity and limited antitumor activity. IT but not SC administration demonstrated systemic immune effects.

{"title":"Intratumoral or Subcutaneous MK-2118, a Non-Cyclic Dinucleotide STING Agonist, With or Without Pembrolizumab for Advanced or Metastatic Solid Tumors or Lymphomas.","authors":"Jason J Luke, Randy F Sweis, J Randolph Hecht, Reva Schneider, Mark N Stein, Talia Golan, Timothy A Yap, Anuradha Khilnani, Mo Huang, Runchen Zhao, Thomas Jemielita, Sandip Pravin Patel","doi":"10.1158/1078-0432.CCR-24-2824","DOIUrl":"https://doi.org/10.1158/1078-0432.CCR-24-2824","url":null,"abstract":"<p><strong>Background: </strong>We evaluated the non-cyclic dinucleotide stimulator of interferon genes agonist MK-2118 ± pembrolizumab in patients with advanced solid tumors or lymphomas.</p><p><strong>Methods: </strong>This first-in-human study (NCT03249792) enrolled patients with refractory, advanced solid tumors or lymphomas. Patients received intratumoral (IT) MK-2118 100-20,000 µg (arm 1), IT MK-2118 900-15,000 µg plus intravenous (IV) pembrolizumab 200 mg every 3 weeks (Q3W; arm 2), or subcutaneous (SC) MK-2118 5000-150,000 µg plus IV pembrolizumab 200 mg Q3W (arm 4); arm 3 (visceral injection of MK-2118) was not pursued. IT dosing used an accelerated titration design and modified toxicity probability interval method; SC dosing (arm 4) was started subsequent to arms 1 and 2. Primary objectives were safety/tolerability. MK-2118 pharmacokinetics were a secondary endpoint; objective responses and biomarkers were exploratory endpoints.</p><p><strong>Results: </strong>140 patients were enrolled (arm 1, n=27; arm 2, n=57; arm 4, n=56). Grade 3/4 treatment-related adverse events occurred in 22%, 23%, and 11% of patients, respectively, but no maximum tolerated dose was identified up to MK-2118 20,000, 15,000, and 150,000 µg across the 3 arms. Dose-dependent increases in MK-2118 systemic exposure were observed following IT and SC administration. Objective responses were seen in 0%, 6%, and 4% of patients, respectively. IT MK-2118 led to dose-dependent changes in STING-based blood RNA expression levels, interferon-gamma, interferon-gamma‒induced protein 10, and interleukin-6; SC MK-2118 did not generate dose-related immune responses.</p><p><strong>Conclusion: </strong>IT MK-2118 ± pembrolizumab and SC MK-2118 plus pembrolizumab had manageable toxicity and limited antitumor activity. IT but not SC administration demonstrated systemic immune effects.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":10.0,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022360","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}
引用次数: 0
First-in-Human Phase 1 study of a CD16A bispecific innate cell engager, AFM24, targeting EGFR-expressing solid tumors.
IF 1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-01-23 DOI: 10.1158/1078-0432.CCR-24-1991
Anthony El-Khoueiry, Omar Saavedra, Jacob Thomas, Claire Livings, Elena Garralda, Gabriele Hintzen, Laura Kohlhas, Dessislava Vanosmael, Joachim Koch, Erich Rajkovic, Paulien Ravenstijn, Paolo Nuciforo, Todd A Fehniger, Mark Foster, Melissa M Berrien-Elliott, Susanne Wingert, Sina Stäble, Daniela Morales-Espinosa, Delcia Rivas, Michael Emig, Juanita Lopez

Purpose: Innate immune cell-based therapies have shown promising antitumor activity against solid and hematologic malignancies. AFM24, a bispecific innate cell engager, binds CD16A on natural killer (NK) cells/macrophages and EGFR on tumor cells, redirecting antitumor activity towards tumors. The safety and tolerability of AFM24 was evaluated in this Phase 1/2a dose escalation/dose expansion study in patients with recurrent or persistent, advanced solid tumors known to express EGFR.

Methods: The main objective in Phase 1 was to determine the maximum tolerated dose (MTD) and/or recommended phase 2 dose (RP2D). The primary endpoint was the incidence of dose-limiting toxicities (DLTs) during the observation period. Secondary endpoints included the incidence of treatment emergent adverse events and pharmacokinetics.

Results: In the dose escalation phase, 35 patients received AFM24 weekly across seven dose cohorts (14-720 mg). One patient experienced a DLT of Grade 3 infusion-related reaction (IRR). IRRs were mainly reported after the first infusion; these were manageable with premedication and a gradual increase in infusion rate. Pharmacokinetics were dose-proportional and CD16A receptor occupancy on NK cells approached saturation between 320-480 mg. Paired tumor biopsies demonstrated activation of innate and adaptive immune responses within the tumor. Best objective response was stable disease in 10/35 patients; four had stable disease for 4.3-7.1 months.

Conclusions: AFM24 was well tolerated with 480 mg established as the RP2D. AFM24 could be a novel therapy for patients with EGFR-expressing solid tumors with suitable tolerability and appropriate pharmacokinetic properties for further development in combination with other immuno-oncology therapeutics.

{"title":"First-in-Human Phase 1 study of a CD16A bispecific innate cell engager, AFM24, targeting EGFR-expressing solid tumors.","authors":"Anthony El-Khoueiry, Omar Saavedra, Jacob Thomas, Claire Livings, Elena Garralda, Gabriele Hintzen, Laura Kohlhas, Dessislava Vanosmael, Joachim Koch, Erich Rajkovic, Paulien Ravenstijn, Paolo Nuciforo, Todd A Fehniger, Mark Foster, Melissa M Berrien-Elliott, Susanne Wingert, Sina Stäble, Daniela Morales-Espinosa, Delcia Rivas, Michael Emig, Juanita Lopez","doi":"10.1158/1078-0432.CCR-24-1991","DOIUrl":"https://doi.org/10.1158/1078-0432.CCR-24-1991","url":null,"abstract":"<p><strong>Purpose: </strong>Innate immune cell-based therapies have shown promising antitumor activity against solid and hematologic malignancies. AFM24, a bispecific innate cell engager, binds CD16A on natural killer (NK) cells/macrophages and EGFR on tumor cells, redirecting antitumor activity towards tumors. The safety and tolerability of AFM24 was evaluated in this Phase 1/2a dose escalation/dose expansion study in patients with recurrent or persistent, advanced solid tumors known to express EGFR.</p><p><strong>Methods: </strong>The main objective in Phase 1 was to determine the maximum tolerated dose (MTD) and/or recommended phase 2 dose (RP2D). The primary endpoint was the incidence of dose-limiting toxicities (DLTs) during the observation period. Secondary endpoints included the incidence of treatment emergent adverse events and pharmacokinetics.</p><p><strong>Results: </strong>In the dose escalation phase, 35 patients received AFM24 weekly across seven dose cohorts (14-720 mg). One patient experienced a DLT of Grade 3 infusion-related reaction (IRR). IRRs were mainly reported after the first infusion; these were manageable with premedication and a gradual increase in infusion rate. Pharmacokinetics were dose-proportional and CD16A receptor occupancy on NK cells approached saturation between 320-480 mg. Paired tumor biopsies demonstrated activation of innate and adaptive immune responses within the tumor. Best objective response was stable disease in 10/35 patients; four had stable disease for 4.3-7.1 months.</p><p><strong>Conclusions: </strong>AFM24 was well tolerated with 480 mg established as the RP2D. AFM24 could be a novel therapy for patients with EGFR-expressing solid tumors with suitable tolerability and appropriate pharmacokinetic properties for further development in combination with other immuno-oncology therapeutics.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":10.0,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143022357","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}
引用次数: 0
期刊
Clinical Cancer Research
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