Pub Date : 2025-12-02DOI: 10.1158/1078-0432.CCR-25-3784
Youngho Seo, Robert R Flavell
Hepatocellular carcinoma (HCC) lacks precise and noninvasive diagnostic tools. To address this a new positron emission tomography (PET) radiotracer, [68Ga]Ga-XH-06, was developed. It uses an antibody fragment to target glypican-3 (GPC3), a highly specific biomarker, successfully visualizing GPC3 expressions in patients with HCC, potentially meeting the important unmet need.
{"title":"Clinical translation of antibody positron emission tomography for cancer imaging.","authors":"Youngho Seo, Robert R Flavell","doi":"10.1158/1078-0432.CCR-25-3784","DOIUrl":"https://doi.org/10.1158/1078-0432.CCR-25-3784","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) lacks precise and noninvasive diagnostic tools. To address this a new positron emission tomography (PET) radiotracer, [68Ga]Ga-XH-06, was developed. It uses an antibody fragment to target glypican-3 (GPC3), a highly specific biomarker, successfully visualizing GPC3 expressions in patients with HCC, potentially meeting the important unmet need.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":10.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653899","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-02DOI: 10.1158/1078-0432.CCR-25-0434
Maria Armero, Manuel Smolkin, Mario Garcia-Dominguez, Pedro Berraondo, Ignacio Melero, Felipe Galvez-Cancino
Monoclonal antibodies (mAbs) used in the clinic can engage the immune system through their Fc domains, eliciting effector mechanisms such as, antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP). While natural killer (NK) cells have historically been implicated as the principal mediators of ADCC, emerging evidence suggests that tumour-associated macrophages (TAMs), particularly monocyte-derived subsets, play a central role in ADCP. These macrophages are abundant within the tumour microenvironment, express high levels of activating Fc gamma receptors (FcγRs), and retain robust phagocytic capacity. However, the heterogeneity of TAMs and the limited understanding of FcγR expression patterns across tumour types have constrained the therapeutic exploitation of ADCP. This review critically examines the contribution of macrophage-mediated ADCP to the activity of both tumour-targeting and immune-regulatory mAbs. We discuss how FcγR polymorphisms, isotype engineering, and antibody effector functions influence therapeutic efficacy, with particular attention to commonly used tumour targeting and immunomodulatory antibodies. The role of inhibitory receptors such as FcγRIIb, CD47, and PD-1 in modulating ADCP is also addressed. Advances in multispecific antibodies targeting tumour antigens alongside ADCP regulators, and preclinical data highlighting the importance of FcγR engagement, underscore the untapped potential of this pathway. We highlight key challenges, including TAM heterogeneity and macrophage hypophagia. Emerging strategies integrating synthetic biology and FcγR profiling may enable the rational design of therapies that selectively enhance ADCP. Macrophage-mediated ADCP represents a promising but underexploited axis in cancer immunotherapy, warranting further mechanistic investigation and translational development.
{"title":"Facts and hopes in harnessing macrophage-mediated antibody-dependent cellular phagocytosis for cancer immunotherapy.","authors":"Maria Armero, Manuel Smolkin, Mario Garcia-Dominguez, Pedro Berraondo, Ignacio Melero, Felipe Galvez-Cancino","doi":"10.1158/1078-0432.CCR-25-0434","DOIUrl":"https://doi.org/10.1158/1078-0432.CCR-25-0434","url":null,"abstract":"<p><p>Monoclonal antibodies (mAbs) used in the clinic can engage the immune system through their Fc domains, eliciting effector mechanisms such as, antibody-dependent cellular cytotoxicity (ADCC), and antibody-dependent cellular phagocytosis (ADCP). While natural killer (NK) cells have historically been implicated as the principal mediators of ADCC, emerging evidence suggests that tumour-associated macrophages (TAMs), particularly monocyte-derived subsets, play a central role in ADCP. These macrophages are abundant within the tumour microenvironment, express high levels of activating Fc gamma receptors (FcγRs), and retain robust phagocytic capacity. However, the heterogeneity of TAMs and the limited understanding of FcγR expression patterns across tumour types have constrained the therapeutic exploitation of ADCP. This review critically examines the contribution of macrophage-mediated ADCP to the activity of both tumour-targeting and immune-regulatory mAbs. We discuss how FcγR polymorphisms, isotype engineering, and antibody effector functions influence therapeutic efficacy, with particular attention to commonly used tumour targeting and immunomodulatory antibodies. The role of inhibitory receptors such as FcγRIIb, CD47, and PD-1 in modulating ADCP is also addressed. Advances in multispecific antibodies targeting tumour antigens alongside ADCP regulators, and preclinical data highlighting the importance of FcγR engagement, underscore the untapped potential of this pathway. We highlight key challenges, including TAM heterogeneity and macrophage hypophagia. Emerging strategies integrating synthetic biology and FcγR profiling may enable the rational design of therapies that selectively enhance ADCP. Macrophage-mediated ADCP represents a promising but underexploited axis in cancer immunotherapy, warranting further mechanistic investigation and translational development.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":10.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653955","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-02DOI: 10.1158/1078-0432.ccr-25-2819
Vivek Podder,Robert L Coleman,Andrea R Hagemann,Priya Singhania,Matthew A Powell,Thomas J Herzog,Brian M Slomovitz
Endometrial cancer (EC) incidence and mortality is rising globally, largely driven by the obesity epidemic. Treatment options remain limited for obesity-associated, hormone-resistant, or fertility-preserving EC, highlighting the need for novel therapies that address both tumor biology and metabolic dysfunction. Here, we synthesize the molecular rationale, preclinical, population-based, and emerging clinical trial evidence on glucagon-like peptide-1 receptor (GLP-1R) agonists (GLP-1 RAs) in EC. GLP-1R is expressed in benign and malignant endometrial tissues, where activation of cAMP-PKA and AMPK-mTOR signaling has been shown to mediate antiproliferative, proapoptotic, and autophagy-inducing effects. Preclinical models demonstrate that class-wide GLP-1 RAs may restore progesterone receptor expression, overcome hormone resistance, and synergize with progestin therapy; however, the effects may vary by agent. Retrospective studies suggest that combining GLP-1 RAs with local progestin therapy, commonly a levonorgestrel-releasing intrauterine device (preferred in obesity because oral progestin bioavailability is reduced), is associated with reduced EC in high-risk women. Ongoing clinical trials are assessing their role in fertility-sparing settings-evaluating complete response to progestin-based therapy, relapse rates, and time to conception-as well as in adjuvant settings, where disease-free survival is a key endpoint; however, gastrointestinal tolerability and the absence of long-term safety data in EC populations remain important considerations. As a class, GLP-1 RAs represent a promising therapeutic approach to targeting both the obesogenic milieu and tumor-intrinsic pathways in EC; however, agent-specific differences warrant attention. Prospective, subtype-stratified trials are essential to establish their role in comprehensive EC care.
{"title":"Repositioning GLP-1 Receptor Agonists in Endometrial Cancer: Molecular Rationale, Preclinical Insights, and Translational Opportunities.","authors":"Vivek Podder,Robert L Coleman,Andrea R Hagemann,Priya Singhania,Matthew A Powell,Thomas J Herzog,Brian M Slomovitz","doi":"10.1158/1078-0432.ccr-25-2819","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-25-2819","url":null,"abstract":"Endometrial cancer (EC) incidence and mortality is rising globally, largely driven by the obesity epidemic. Treatment options remain limited for obesity-associated, hormone-resistant, or fertility-preserving EC, highlighting the need for novel therapies that address both tumor biology and metabolic dysfunction. Here, we synthesize the molecular rationale, preclinical, population-based, and emerging clinical trial evidence on glucagon-like peptide-1 receptor (GLP-1R) agonists (GLP-1 RAs) in EC. GLP-1R is expressed in benign and malignant endometrial tissues, where activation of cAMP-PKA and AMPK-mTOR signaling has been shown to mediate antiproliferative, proapoptotic, and autophagy-inducing effects. Preclinical models demonstrate that class-wide GLP-1 RAs may restore progesterone receptor expression, overcome hormone resistance, and synergize with progestin therapy; however, the effects may vary by agent. Retrospective studies suggest that combining GLP-1 RAs with local progestin therapy, commonly a levonorgestrel-releasing intrauterine device (preferred in obesity because oral progestin bioavailability is reduced), is associated with reduced EC in high-risk women. Ongoing clinical trials are assessing their role in fertility-sparing settings-evaluating complete response to progestin-based therapy, relapse rates, and time to conception-as well as in adjuvant settings, where disease-free survival is a key endpoint; however, gastrointestinal tolerability and the absence of long-term safety data in EC populations remain important considerations. As a class, GLP-1 RAs represent a promising therapeutic approach to targeting both the obesogenic milieu and tumor-intrinsic pathways in EC; however, agent-specific differences warrant attention. Prospective, subtype-stratified trials are essential to establish their role in comprehensive EC care.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"6 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145657050","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1158/1078-0432.ccr-24-4295
Keun-Wook Lee, Li-Yuan Bai, Minkyu Jung, Jieer Ying, Young-Hyuck Im, Do-Youn Oh, Jae Yong Cho, Sang Cheul Oh, Yee Chao, Jin Won Kim, Ye Chen, Vincent Li, Shengnan Chen, Yoon-Koo Kang
Purpose: This phase 1b/2 trial (NCT04276493) assessed the antitumor activity, safety and pharmacokinetics (PK) of zanidatamab in combination with tislelizumab and chemotherapy in patients with advanced HER2-positive (HER2+) gastric/gastroesophageal junction cancer (GC/GEJC). Patients and Methods: Adult patients with previously untreated, unresectable, locally advanced/metastatic HER2+ GC/GEJC received zanidatamab 30 mg/kg intravenously (IV) (cohort A) or zanidatamab 1800 mg IV (weight <70 kg)/2400 mg IV (weight ≥70 kg) (cohort B) once every 3 weeks (Q3W). Both cohorts received tislelizumab 200 mg IV Q3W and standard chemotherapy (capecitabine and oxaliplatin [CAPOX]) Q3W. Primary endpoints were investigator-assessed confirmed objective response rate (cORR) per RECIST v1.1, in addition to frequency and severity of adverse events (AEs) and serious AEs. Secondary endpoints included investigator-assessed progression-free survival (PFS), duration of response (DoR), overall survival (OS), PK, and immunogenicity of zanidatamab. Results: As of December 7, 2023, 33 patients (cohort A, n=19; cohort B, n=14) received treatment. cORR was 75.8%; median DoR, PFS, and OS were 23.3, 16.7, and 32.4 months, respectively. The most common treatment-related AEs (TRAEs) were diarrhea (100%), nausea (63.6%), and decreased appetite (48.5%). TRAEs of grade ≥3 were reported in 22 (66.7%) patients; diarrhea was the most common (27.3%). Conclusions: Zanidatamab, in combination with tislelizumab and CAPOX, demonstrated clinically meaningful antitumor activity with a manageable safety profile as first-line therapy for patients with HER2+ GC/GEJC. These results support further development of zanidatamab and tislelizumab with chemotherapy in this patient population in the ongoing phase 3 HERIZON-GEA-01 trial (NCT05152147).
{"title":"Phase 1b/2 Study of Zanidatamab in Combination with Tislelizumab and Chemotherapy in First-Line HER2-Positive Gastric/Gastroesophageal Junction Adenocarcinoma","authors":"Keun-Wook Lee, Li-Yuan Bai, Minkyu Jung, Jieer Ying, Young-Hyuck Im, Do-Youn Oh, Jae Yong Cho, Sang Cheul Oh, Yee Chao, Jin Won Kim, Ye Chen, Vincent Li, Shengnan Chen, Yoon-Koo Kang","doi":"10.1158/1078-0432.ccr-24-4295","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-24-4295","url":null,"abstract":"Purpose: This phase 1b/2 trial (NCT04276493) assessed the antitumor activity, safety and pharmacokinetics (PK) of zanidatamab in combination with tislelizumab and chemotherapy in patients with advanced HER2-positive (HER2+) gastric/gastroesophageal junction cancer (GC/GEJC). Patients and Methods: Adult patients with previously untreated, unresectable, locally advanced/metastatic HER2+ GC/GEJC received zanidatamab 30 mg/kg intravenously (IV) (cohort A) or zanidatamab 1800 mg IV (weight &lt;70 kg)/2400 mg IV (weight ≥70 kg) (cohort B) once every 3 weeks (Q3W). Both cohorts received tislelizumab 200 mg IV Q3W and standard chemotherapy (capecitabine and oxaliplatin [CAPOX]) Q3W. Primary endpoints were investigator-assessed confirmed objective response rate (cORR) per RECIST v1.1, in addition to frequency and severity of adverse events (AEs) and serious AEs. Secondary endpoints included investigator-assessed progression-free survival (PFS), duration of response (DoR), overall survival (OS), PK, and immunogenicity of zanidatamab. Results: As of December 7, 2023, 33 patients (cohort A, n=19; cohort B, n=14) received treatment. cORR was 75.8%; median DoR, PFS, and OS were 23.3, 16.7, and 32.4 months, respectively. The most common treatment-related AEs (TRAEs) were diarrhea (100%), nausea (63.6%), and decreased appetite (48.5%). TRAEs of grade ≥3 were reported in 22 (66.7%) patients; diarrhea was the most common (27.3%). Conclusions: Zanidatamab, in combination with tislelizumab and CAPOX, demonstrated clinically meaningful antitumor activity with a manageable safety profile as first-line therapy for patients with HER2+ GC/GEJC. These results support further development of zanidatamab and tislelizumab with chemotherapy in this patient population in the ongoing phase 3 HERIZON-GEA-01 trial (NCT05152147).","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"30 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145651229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1158/1078-0432.CCR-25-1628
Amanda M Shafer, Emma Kenna, Lexi-Ann F Golden, Ahmed M Elhossiny, Kyle D Perry, Jodi Wilkowski, Wei Yan, Brynn Kaczkofsky, Jake McGue, Scott C Bresler, Adam H Courtney, Jessie M Dalman, Craig J Galbán, Wei Jiang, Carlos E Espinoza, Rashmi Chugh, Matthew K Iyer, Timothy L Frankel, Marina Pasca di Magliano, Andrzej A Dlugosz, Christina V Angeles
Purpose: Liposarcoma is the most common soft tissue sarcoma. Well-differentiated liposarcoma can progress to dedifferentiated liposarcoma (DDLPS), a more aggressive form with higher metastatic potential and poor response to existing therapies. Progress in understanding and treating liposarcoma has been limited. To address this, we sought to develop an immunocompetent genetically engineered mouse model of liposarcoma.
Experimental design: We developed an autochthonous, immunocompetent liposarcoma mouse model [AAV8-Ap2.2-eGFP/Cre Ptenf/f;Trp53f/f C57BL/6 (ACPP)] by using targeted Cre-mediated deletion of Trp53 and Pten in adipocytes to mimic the signaling alterations observed in human liposarcoma. We characterized the histology, transcriptional features, and tumor microenvironment of this model. Additionally, we established syngeneic cell lines derived from ACPP DDLPS tumors and evaluated them for tumor formation, growth dynamics, and immune composition after implantation.
Results: ACPP mice develop well-differentiated liposarcoma, DDLPS, and mixed tumors, mirroring human disease. Both murine and human DDLPS tumors share key transcriptional features and exhibit heterogeneous T-cell infiltration. Syngeneic DDLPS cell lines reliably form tumors in vivo, with each line demonstrating distinct growth kinetics, aggressiveness, and immune profiles.
Conclusions: The ACPP model provides a novel and clinically relevant platform to study liposarcoma in an immunocompetent setting. Along with the ACPP-derived cell lines, these models not only provide essential tools to understand the complex immunobiology of liposarcoma but also can be used to elucidate the underlying molecular mechanisms driving liposarcoma generation and progression and significantly accelerate the pace of preclinical studies aimed at uncovering more effective new therapies for patients with this aggressive malignancy.
{"title":"Immunocompetent Murine Models Recapitulate the Heterogeneous Tumor-Immune Microenvironment of Human Liposarcoma.","authors":"Amanda M Shafer, Emma Kenna, Lexi-Ann F Golden, Ahmed M Elhossiny, Kyle D Perry, Jodi Wilkowski, Wei Yan, Brynn Kaczkofsky, Jake McGue, Scott C Bresler, Adam H Courtney, Jessie M Dalman, Craig J Galbán, Wei Jiang, Carlos E Espinoza, Rashmi Chugh, Matthew K Iyer, Timothy L Frankel, Marina Pasca di Magliano, Andrzej A Dlugosz, Christina V Angeles","doi":"10.1158/1078-0432.CCR-25-1628","DOIUrl":"10.1158/1078-0432.CCR-25-1628","url":null,"abstract":"<p><strong>Purpose: </strong>Liposarcoma is the most common soft tissue sarcoma. Well-differentiated liposarcoma can progress to dedifferentiated liposarcoma (DDLPS), a more aggressive form with higher metastatic potential and poor response to existing therapies. Progress in understanding and treating liposarcoma has been limited. To address this, we sought to develop an immunocompetent genetically engineered mouse model of liposarcoma.</p><p><strong>Experimental design: </strong>We developed an autochthonous, immunocompetent liposarcoma mouse model [AAV8-Ap2.2-eGFP/Cre Ptenf/f;Trp53f/f C57BL/6 (ACPP)] by using targeted Cre-mediated deletion of Trp53 and Pten in adipocytes to mimic the signaling alterations observed in human liposarcoma. We characterized the histology, transcriptional features, and tumor microenvironment of this model. Additionally, we established syngeneic cell lines derived from ACPP DDLPS tumors and evaluated them for tumor formation, growth dynamics, and immune composition after implantation.</p><p><strong>Results: </strong>ACPP mice develop well-differentiated liposarcoma, DDLPS, and mixed tumors, mirroring human disease. Both murine and human DDLPS tumors share key transcriptional features and exhibit heterogeneous T-cell infiltration. Syngeneic DDLPS cell lines reliably form tumors in vivo, with each line demonstrating distinct growth kinetics, aggressiveness, and immune profiles.</p><p><strong>Conclusions: </strong>The ACPP model provides a novel and clinically relevant platform to study liposarcoma in an immunocompetent setting. Along with the ACPP-derived cell lines, these models not only provide essential tools to understand the complex immunobiology of liposarcoma but also can be used to elucidate the underlying molecular mechanisms driving liposarcoma generation and progression and significantly accelerate the pace of preclinical studies aimed at uncovering more effective new therapies for patients with this aggressive malignancy.</p>","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":" ","pages":"5078-5095"},"PeriodicalIF":10.2,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12664306/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145032829","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
PURPOSEThe optimal conversion treatment for patients with borderline resectable pancreatic cancer or locally advanced pancreatic cancer (BRPC/LAPC) remains unclear. Here we present the efficacy and safety results of a phase II trial evaluating tislelizumab combined with hypofractionated radiotherapy plus nab-paclitaxel/gemcitabine (THAG) in BRPC/LAPC patients (ChiCTR2000032955, NCT05634564).PATIENTS AND METHODSThis phase II trial enrolled 56 BRPC/LAPC patients (BRPC: 17, 30.4%; LAPC: 39, 69.6%). Participants received tislelizumab plus AG (nab-paclitaxel/gemcitabine) in 21-day cycles. Non-progressing patients received concurrent radiotherapy during the third chemotherapy cycle. After four treatment cycles, a multidisciplinary team (MDT) assessed eligibility for radical surgery. Dynamic biomolecular profiling was performed.RESULTSFifty-six eligible patients were enrolled. The objective response rate (ORR) was 51.8% (95% CI 38.0-65.3%). Median progression-free survival (mPFS) was 13.2 months (95% CI 11.6-19.4 months) and median overall survival (mOS) was 21.3 months (95% CI 18.8-not reached [NR]). Among 30 patients who reached criteria for surgical resectability, 22 patients (22/56, 39.3%) underwent radical resection, comprising 9 BRPC patients (9/17, 52.9%) and 13 LAPC patients (13/39, 33.3%). The R0 resection rate reached 90.9% (95% CI 70.8-98.9%), and the mOS of patients who underwent surgery was 34.0 months (95%CI 20.1-NR). Grade ≥3 adverse events (AEs) occurred in 33/56 patients (58.9%). Dynamic biomarker exploration revealed that baseline IL-6 level (>5 pg/ml) predicted better PFS. Moreover, ctDNA status and clearance demonstrated superior survival.CONCLUSIONSThe THAG regimen as preoperative therapy showed encouraging clinical activity with a manageable safety profile. Dynamic biomarker findings reveal potential for guiding precision treatment strategies with THAG.
目的边缘可切除胰腺癌或局部晚期胰腺癌(BRPC/LAPC)患者的最佳转化治疗尚不清楚。在此,我们提出了一项II期试验的有效性和安全性结果,该试验评估了tislelizumab联合低分割放疗加nab-紫杉醇/吉西他滨(THAG)治疗BRPC/LAPC患者(ChiCTR2000032955, NCT05634564)。患者和方法该II期试验纳入了56例BRPC/LAPC患者(BRPC: 17例,30.4%;LAPC: 39例,69.6%)。参与者接受tislelizumab加AG (nab-紫杉醇/吉西他滨)治疗,周期为21天。无进展患者在第三个化疗周期内接受同步放疗。四个治疗周期后,一个多学科小组(MDT)评估根治性手术的资格。进行动态生物分子分析。结果入选56例符合条件的患者。客观有效率(ORR)为51.8% (95% CI 38.0 ~ 65.3%)。中位无进展生存期(mPFS)为13.2个月(95% CI 11.6-19.4个月),中位总生存期(mOS)为21.3个月(95% CI 18.8-未达到[NR])。30例达到手术可切除标准的患者中,22例(22/56,39.3%)行根治性切除,其中BRPC 9例(9/17,52.9%),LAPC 13例(13/39,33.3%)。R0切除率达90.9% (95%CI 70.8 ~ 98.9%),手术患者的生存期为34.0个月(95%CI 20.1 ~ nr)。56例患者中有33例(58.9%)发生≥3级不良事件(ae)。动态生物标志物探索显示,基线IL-6水平(bbb50 pg/ml)预测更好的PFS。此外,ctDNA状态和清除率显示出更高的生存率。结论THAG方案作为术前治疗具有良好的临床活性和可管理的安全性。动态生物标志物的发现揭示了指导THAG精确治疗策略的潜力。
{"title":"Tislelizumab and hypofractionated radiotherapy plus nab-paclitaxel/gemcitabine as conversion therapy for BRPC/LAPC: A Phase II trial with dynamic biomarker monitoring.","authors":"Jiayao Ni,Yi Sun,Hao Cheng,Xinyuan Bai,Fan Tong,Xing Zhang,Xiaoxuan Wang,Liang Mao,Gang Li,Shanshan Shen,Weiwei Kong,Jian He,Aimei Li,Jun Chen,Min Tang,Bozhu Chen,Qi Li,Qing Gu,Zhongyu Lu,Dongsheng Chen,Huizi Sha,Baorui Liu,Yudong Qiu,Juan Du","doi":"10.1158/1078-0432.ccr-25-2461","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-25-2461","url":null,"abstract":"PURPOSEThe optimal conversion treatment for patients with borderline resectable pancreatic cancer or locally advanced pancreatic cancer (BRPC/LAPC) remains unclear. Here we present the efficacy and safety results of a phase II trial evaluating tislelizumab combined with hypofractionated radiotherapy plus nab-paclitaxel/gemcitabine (THAG) in BRPC/LAPC patients (ChiCTR2000032955, NCT05634564).PATIENTS AND METHODSThis phase II trial enrolled 56 BRPC/LAPC patients (BRPC: 17, 30.4%; LAPC: 39, 69.6%). Participants received tislelizumab plus AG (nab-paclitaxel/gemcitabine) in 21-day cycles. Non-progressing patients received concurrent radiotherapy during the third chemotherapy cycle. After four treatment cycles, a multidisciplinary team (MDT) assessed eligibility for radical surgery. Dynamic biomolecular profiling was performed.RESULTSFifty-six eligible patients were enrolled. The objective response rate (ORR) was 51.8% (95% CI 38.0-65.3%). Median progression-free survival (mPFS) was 13.2 months (95% CI 11.6-19.4 months) and median overall survival (mOS) was 21.3 months (95% CI 18.8-not reached [NR]). Among 30 patients who reached criteria for surgical resectability, 22 patients (22/56, 39.3%) underwent radical resection, comprising 9 BRPC patients (9/17, 52.9%) and 13 LAPC patients (13/39, 33.3%). The R0 resection rate reached 90.9% (95% CI 70.8-98.9%), and the mOS of patients who underwent surgery was 34.0 months (95%CI 20.1-NR). Grade ≥3 adverse events (AEs) occurred in 33/56 patients (58.9%). Dynamic biomarker exploration revealed that baseline IL-6 level (>5 pg/ml) predicted better PFS. Moreover, ctDNA status and clearance demonstrated superior survival.CONCLUSIONSThe THAG regimen as preoperative therapy showed encouraging clinical activity with a manageable safety profile. Dynamic biomarker findings reveal potential for guiding precision treatment strategies with THAG.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"21 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145644940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1158/1078-0432.ccr-25-2890
Michael J Morris,Jessica Flynn,Binsheng Zhao,Aseem Anand,Karl Sjöstrand,Steven M Larson,Ashley M Regazzi,Gary V Borzillo,Autumn Park,Lawrence H Schwartz,Glenn Heller
PURPOSEContemporary prostate cancer prognostic models do not include imaging and generally are based on pretreatment parameters. We sought to develop an externally validated model that used novel quantification of soft-tissue and bone disease, integrated with standard clinical and serum biomarkers, at baseline and up to 6 months of treatment.PATIENTS AND METHODSTwo randomized phase 3 trials, Cougar COU-AA-302 (NCT00887198; for derivation) and Alliance A031201 (NCT01949337; for validation), were used to evaluate the added value of early on-treatment bone imaging and over 1000 radiomics features on CT, used in conjunction with clinical and serum biomarkers in first-line metastatic castration-resistant prostate cancer. Predictive accuracy measures were computed to determine whether these early on-treatment biomarkers could reliably sort patients into risk groups that inform overall survival and whether the patient-specific biomarker risk score could precisely predict their overall survival time.RESULTSImaging improved patient risk stratification but did not improve individual survival predictions. The strongest risk prediction model was developed for patients with bone-only metastases. This model was also the least complex, relying on just 16 risk factors, whereas all other models were high-dimensional, incorporating approximately 1100 intercept and 1100 slope features from the early on-treatment biomarker trajectories.CONCLUSIONSPretreatment and early on-treatment serum and automated quantitative imaging markers can well discriminate risk of death. Imaging improves this risk categorization relative to serum biomarkers alone. Such models can give early outcome predictions and can be used in future trials that involve imaging, even using traditional techniques such as bone scintigraphy.
目的:继发性前列腺癌预后模型不包括影像学,通常基于预处理参数。我们试图开发一种外部验证的模型,该模型在基线和治疗6个月时使用新的软组织和骨骼疾病量化,并与标准临床和血清生物标志物相结合。患者和方法两项随机3期试验,Cougar cu - aa -302 (NCT00887198;用于衍生)和Alliance A031201 (NCT01949337;用于验证),用于评估早期治疗骨成像和超过1000个CT放射组学特征的附加价值,与临床和血清生物标志物一起用于一线转移性去势抵抗性前列腺癌。计算预测准确性测量,以确定这些早期治疗生物标志物是否能够可靠地将患者分类到告知总体生存的风险组,以及患者特异性生物标志物风险评分是否能够准确预测其总体生存时间。结果成像改善了患者的风险分层,但没有改善个体生存预测。仅骨转移患者的风险预测模型最强。该模型也是最不复杂的,仅依赖于16个风险因素,而所有其他模型都是高维的,包含了来自早期治疗生物标志物轨迹的大约1100个截距和1100个斜率特征。结论治疗前和治疗早期血清和自动定量影像学指标能较好地区分死亡风险。相对于单独的血清生物标志物,影像学改善了这种风险分类。这些模型可以提供早期结果预测,并可用于涉及成像的未来试验,甚至可以使用骨闪烁成像等传统技术。
{"title":"Automated imaging as an adjunct to serum and clinical biomarkers: a new validated prediction tool for metastatic castration-resistant prostate cancer.","authors":"Michael J Morris,Jessica Flynn,Binsheng Zhao,Aseem Anand,Karl Sjöstrand,Steven M Larson,Ashley M Regazzi,Gary V Borzillo,Autumn Park,Lawrence H Schwartz,Glenn Heller","doi":"10.1158/1078-0432.ccr-25-2890","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-25-2890","url":null,"abstract":"PURPOSEContemporary prostate cancer prognostic models do not include imaging and generally are based on pretreatment parameters. We sought to develop an externally validated model that used novel quantification of soft-tissue and bone disease, integrated with standard clinical and serum biomarkers, at baseline and up to 6 months of treatment.PATIENTS AND METHODSTwo randomized phase 3 trials, Cougar COU-AA-302 (NCT00887198; for derivation) and Alliance A031201 (NCT01949337; for validation), were used to evaluate the added value of early on-treatment bone imaging and over 1000 radiomics features on CT, used in conjunction with clinical and serum biomarkers in first-line metastatic castration-resistant prostate cancer. Predictive accuracy measures were computed to determine whether these early on-treatment biomarkers could reliably sort patients into risk groups that inform overall survival and whether the patient-specific biomarker risk score could precisely predict their overall survival time.RESULTSImaging improved patient risk stratification but did not improve individual survival predictions. The strongest risk prediction model was developed for patients with bone-only metastases. This model was also the least complex, relying on just 16 risk factors, whereas all other models were high-dimensional, incorporating approximately 1100 intercept and 1100 slope features from the early on-treatment biomarker trajectories.CONCLUSIONSPretreatment and early on-treatment serum and automated quantitative imaging markers can well discriminate risk of death. Imaging improves this risk categorization relative to serum biomarkers alone. Such models can give early outcome predictions and can be used in future trials that involve imaging, even using traditional techniques such as bone scintigraphy.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"198200 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145644896","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}
BACKGROUNDHER2DX is a validated genomic assay used to support treatment decisions in early-stage HER2-positive (HER2+) breast cancer. It provides three scores: relapse risk, likelihood of pathologic complete response (pCR), and ERBB2 mRNA expression. This study aimed to evaluate the association between HER2DX and histopathologic features and to assess its relationship with pCR after neoadjuvant therapy.METHODSPatients with newly diagnosed stage I-III HER2+ breast cancer were analyzed based on available HER2DX results during routine care in Spain (January 2022-June 2025). Centralized HER2DX testing was performed on formalin-fixed, paraffin-embedded tumor samples. Histopathologic analysis included tumor grade, hormone receptor (HR) status, histological subtype, Ki67 index, HER2 immunohistochemistry (IHC) score, stromal tumor-infiltrating lymphocytes (TILs), tertiary lymphoid structures (TLS), and spatial immune distribution. Univariate and multivariable logistic regression analyses were conducted to identify factors associated with pCR after neoadjuvant trastuzumab-based therapy.RESULTSA total of 410 HER2+ tumors were analyzed, and 250 patients received neoadjuvant trastuzumab-based therapy with available surgical outcomes (36.0% achieved a pCR). HER2DX pCR scores were significantly associated with all eight histopathologic features, whereas relapse risk and ERBB2 scores were associated with five and two, respectively. TILs correlated with the immune/immunoglobulin (IGG) signature (r=0.59), and Ki67 with the proliferation signature (r=0.50). The HER2DX pCR score remained the only independent predictor of pCR in multivariable analysis (odds ratio [OR]=1.77, 95% CI 1.08-2.97, p=0.030).CONCLUSIONSHER2DX reflects key biological and pathological features of HER2+ breast cancer and independently predicts pCR, supporting its utility for individualized treatment decision-making.
背景:her2dx是一种经过验证的基因组分析方法,用于支持早期HER2阳性(HER2+)乳腺癌的治疗决策。它提供三个评分:复发风险、病理完全缓解可能性(pCR)和ERBB2 mRNA表达。本研究旨在评估HER2DX与新辅助治疗后组织病理特征的关系,并评估其与pCR的关系。方法对西班牙(2022年1月- 2025年6月)新诊断的I-III期HER2+乳腺癌患者进行常规护理中可用的HER2DX结果分析。对福尔马林固定、石蜡包埋的肿瘤标本进行集中HER2DX检测。组织病理学分析包括肿瘤分级、激素受体(HR)状态、组织学亚型、Ki67指数、HER2免疫组化(IHC)评分、间质肿瘤浸润淋巴细胞(TILs)、三级淋巴组织结构(TLS)和免疫空间分布。进行单因素和多因素logistic回归分析,以确定新辅助曲妥珠单抗治疗后与pCR相关的因素。结果共分析了410例HER2+肿瘤,其中250例患者接受了基于曲妥珠单抗的新辅助治疗,手术效果良好(36.0%达到pCR)。HER2DX pCR评分与所有8个组织病理学特征显著相关,而复发风险和ERBB2评分分别与5和2相关。TILs与免疫/免疫球蛋白(IGG)特征相关(r=0.59), Ki67与增殖特征相关(r=0.50)。HER2DX pCR评分仍然是多变量分析中pCR的唯一独立预测因子(优势比[OR]=1.77, 95% CI 1.08-2.97, p=0.030)。结论sher2dx反映了HER2+乳腺癌的关键生物学和病理特征,并能独立预测pCR,支持其在个体化治疗决策中的应用。
{"title":"Associations of the HER2DX Genomic Test with Biological and Pathological Features in HER2-positive Breast Cancer.","authors":"Esther Sanfeliu,Anabel Martínez-Romero,Mercedes Marín-Aguilera,Sandra Cobo,Blanca González-Farré,Eva Hernandez-Illan,Pedro Jares,Joan-Anton Antón Puig-Butillé,Montserrat Muñoz,Raquel Gómez-Bravo,Marta Tapia,Cristina Tebar,Cristina Saura,Santiago Escribà,Jesús Soberino,Javier Cortés,Serafin Morales,Kepa Amillano,Laia Paré,Patricia Villagrasa,Wesley Buckingham,Francisco Pardo,Joel S Parker,Fara Brasó-Maristany,Eva Ciruelos,Rodrigo Sánchez-Bayona,Olga Martinez-Sáez,Juan Miguel Cejalvo,Aleix Prat","doi":"10.1158/1078-0432.ccr-25-3123","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-25-3123","url":null,"abstract":"BACKGROUNDHER2DX is a validated genomic assay used to support treatment decisions in early-stage HER2-positive (HER2+) breast cancer. It provides three scores: relapse risk, likelihood of pathologic complete response (pCR), and ERBB2 mRNA expression. This study aimed to evaluate the association between HER2DX and histopathologic features and to assess its relationship with pCR after neoadjuvant therapy.METHODSPatients with newly diagnosed stage I-III HER2+ breast cancer were analyzed based on available HER2DX results during routine care in Spain (January 2022-June 2025). Centralized HER2DX testing was performed on formalin-fixed, paraffin-embedded tumor samples. Histopathologic analysis included tumor grade, hormone receptor (HR) status, histological subtype, Ki67 index, HER2 immunohistochemistry (IHC) score, stromal tumor-infiltrating lymphocytes (TILs), tertiary lymphoid structures (TLS), and spatial immune distribution. Univariate and multivariable logistic regression analyses were conducted to identify factors associated with pCR after neoadjuvant trastuzumab-based therapy.RESULTSA total of 410 HER2+ tumors were analyzed, and 250 patients received neoadjuvant trastuzumab-based therapy with available surgical outcomes (36.0% achieved a pCR). HER2DX pCR scores were significantly associated with all eight histopathologic features, whereas relapse risk and ERBB2 scores were associated with five and two, respectively. TILs correlated with the immune/immunoglobulin (IGG) signature (r=0.59), and Ki67 with the proliferation signature (r=0.50). The HER2DX pCR score remained the only independent predictor of pCR in multivariable analysis (odds ratio [OR]=1.77, 95% CI 1.08-2.97, p=0.030).CONCLUSIONSHER2DX reflects key biological and pathological features of HER2+ breast cancer and independently predicts pCR, supporting its utility for individualized treatment decision-making.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"21 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145644899","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1158/1078-0432.ccr-25-3327
Sebastian Medina,Naoto Tokuyama,Kamal Hammouda,Tilak Pathak,Tuomas Mirtti,Pingfu Fu,Shilpa Gupta,Priti Lal,Howard M Sandler,Rohann Correa,Susan Chafe,Amit Shah,Jason A Efstathiou,Karen Hoffman,Michael Straza,Mark A Hallman,Richard Jordan,Stephanie L Pugh,Christopher J Sweeney,Anant Madabhushi
PURPOSEDocetaxel improves survival in metastatic hormone-sensitive prostate cancer (mHSPC) and high-risk localized disease, but benefits vary substantially among patients. Without predictive biomarkers, clinicians cannot identify patients who will benefit, exposing many to unnecessary toxicity. We developed and validated an artificial intelligence-based pathology image classifier (APIC) to predict docetaxel benefit.PATIENTS AND METHODSWe analyzed digitized H&E-stained biopsy specimens from two phase 3 trials: CHAARTED (286/790 patients with mHSPC) and NRG/RTOG 0521 (350/563 patients with high-risk localized disease). APIC used features capturing tumor-immune spatial interactions and nuclear heterogeneity. We evaluated the predictive value of APIC for docetaxel benefit on overall survival and castration-resistance using Cox proportional hazards with interaction terms.RESULTSIn CHAARTED, APIC-positive patients (56.7%) showed significant overall survival improvement with docetaxel (hazard ratio [HR] 0.52, 95% confidence interval [CI] 0.31-0.85; P = 0.008) and delayed castration-resistance (HR 0.48, 95% CI 0.33-0.71; P < 0.001), while APIC-negative patients (43.3%) showed no benefit (HR 1.31, 95% CI 0.71-2.44; P = 0.39). Treatment-APIC interactions were significant (P = 0.022 and P = 0.031). In NRG/RTOG 0521, APIC-positive patients (44.7%) demonstrated survival benefit (HR 0.49, 95% CI 0.26-0.92; P = 0.023), while APIC-negative patients (55.3%) showed no benefit. Treatment-APIC interaction was significant (P = 0.024). Predictive value remained significant after adjusting for clinical variables. Limitations include retrospective analysis and need for prospective validation.CONCLUSIONSAPIC predicts docetaxel benefit in both metastatic and localized prostate cancer, independent of clinical factors. Validation in triplet therapy with androgen receptor pathway inhibitors is needed.
多西紫杉醇可提高转移性激素敏感性前列腺癌(mHSPC)和高危局限性疾病患者的生存率,但不同患者的获益差异很大。如果没有预测性生物标志物,临床医生就无法确定哪些患者将受益,从而使许多患者暴露于不必要的毒性。我们开发并验证了一种基于人工智能的病理图像分类器(APIC)来预测多西他赛的疗效。患者和方法我们分析了来自两个3期试验的数字化h&e染色活检标本:CHAARTED(286/790例mHSPC患者)和NRG/RTOG 0521(350/563例高风险局部疾病患者)。APIC使用捕获肿瘤免疫空间相互作用和核异质性的特征。我们使用带有相互作用项的Cox比例风险评估APIC对多西他赛总生存率和去势抵抗的预测价值。结果apic阳性患者(56.7%)在多西他赛治疗后总生存率显著提高(风险比[HR] 0.52, 95%可信区间[CI] 0.31 ~ 0.85, P = 0.008),延迟阉割抵抗(风险比[HR] 0.48, 95% CI 0.33 ~ 0.71, P < 0.001), apic阴性患者(43.3%)无获益(风险比[HR] 1.31, 95% CI 0.71 ~ 2.44, P = 0.39)。治疗- apic相互作用显著(P = 0.022和P = 0.031)。在NRG/RTOG 0521中,apic阳性患者(44.7%)表现出生存获益(HR 0.49, 95% CI 0.26-0.92; P = 0.023),而apic阴性患者(55.3%)没有获益。治疗与apic交互作用显著(P = 0.024)。在调整临床变量后,预测值仍然显著。局限性包括回顾性分析和需要前瞻性验证。结论sapic预测多西他赛在转移性和局限性前列腺癌中的获益,独立于临床因素。需要对雄激素受体途径抑制剂的三联体治疗进行验证。
{"title":"A Computational Pathology Model to Predict Docetaxel Benefit in Localized High-Risk and Metastatic Prostate Cancer.","authors":"Sebastian Medina,Naoto Tokuyama,Kamal Hammouda,Tilak Pathak,Tuomas Mirtti,Pingfu Fu,Shilpa Gupta,Priti Lal,Howard M Sandler,Rohann Correa,Susan Chafe,Amit Shah,Jason A Efstathiou,Karen Hoffman,Michael Straza,Mark A Hallman,Richard Jordan,Stephanie L Pugh,Christopher J Sweeney,Anant Madabhushi","doi":"10.1158/1078-0432.ccr-25-3327","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-25-3327","url":null,"abstract":"PURPOSEDocetaxel improves survival in metastatic hormone-sensitive prostate cancer (mHSPC) and high-risk localized disease, but benefits vary substantially among patients. Without predictive biomarkers, clinicians cannot identify patients who will benefit, exposing many to unnecessary toxicity. We developed and validated an artificial intelligence-based pathology image classifier (APIC) to predict docetaxel benefit.PATIENTS AND METHODSWe analyzed digitized H&E-stained biopsy specimens from two phase 3 trials: CHAARTED (286/790 patients with mHSPC) and NRG/RTOG 0521 (350/563 patients with high-risk localized disease). APIC used features capturing tumor-immune spatial interactions and nuclear heterogeneity. We evaluated the predictive value of APIC for docetaxel benefit on overall survival and castration-resistance using Cox proportional hazards with interaction terms.RESULTSIn CHAARTED, APIC-positive patients (56.7%) showed significant overall survival improvement with docetaxel (hazard ratio [HR] 0.52, 95% confidence interval [CI] 0.31-0.85; P = 0.008) and delayed castration-resistance (HR 0.48, 95% CI 0.33-0.71; P < 0.001), while APIC-negative patients (43.3%) showed no benefit (HR 1.31, 95% CI 0.71-2.44; P = 0.39). Treatment-APIC interactions were significant (P = 0.022 and P = 0.031). In NRG/RTOG 0521, APIC-positive patients (44.7%) demonstrated survival benefit (HR 0.49, 95% CI 0.26-0.92; P = 0.023), while APIC-negative patients (55.3%) showed no benefit. Treatment-APIC interaction was significant (P = 0.024). Predictive value remained significant after adjusting for clinical variables. Limitations include retrospective analysis and need for prospective validation.CONCLUSIONSAPIC predicts docetaxel benefit in both metastatic and localized prostate cancer, independent of clinical factors. Validation in triplet therapy with androgen receptor pathway inhibitors is needed.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"110 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145644903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1158/1078-0432.ccr-25-2298
Mafalda Oliveira,Eva Ciruelos,Guillermo Villacampa,Serafin Morales,Javier Salvador Bofill,Vanesa Quiroga,Joaquín Gavilá,Alexandra Cortegoso,Estela Vega,Fernando Henao,Sonia Servitja,Pablo Tolosa,Samyukta Chillara,Milana Bergamino,Fernando Salvador,Mariana Paes Dias,Juan M Ferrero-Cafiero,Xavier Gonzalez Farre,Tomás Pascual,Cristina Saura
PURPOSETo evaluate the safety and feasibility of ipatasertib combined with trastuzumab and pertuzumab as maintenance therapy after first-line treatment in patients with HER2-positive metastatic breast cancer harboring PIK3CA mutations (PIK3CAmut).EXPERIMENTAL DESIGNThis prospective, multicenter, single-arm, phase 1b study evaluated the safety and preliminary efficacy of ipatasertib, an AKT inhibitor, combined with trastuzumab and pertuzumab (HP), with or without endocrine therapy as maintainance therapy, in patients with unresectable locally advanced or metastatic PIK3CAmut, HER2-positive breast cancer following first-line induction chemotherapy and HP.RESULTSSeventeen patients were enrolled, with a median follow-up of 27.7 months. During the dose selection phase, ipatasertib at 400 mg daily (21 days on, 7 days off) with standard HP was established as the recommended phase 2 dose (RP2D). This decision was based on the absence of dose-limiting toxicities in the first six patients treated at this dose during the initial 28-day cycle, which constituted the primary endpoint. Seven (41.2%) patients experienced grade 3 treatment-related adverse events (TRAEs), with diarrhea and nausea being the most frequent. Two (11.8%) reported four serious TRAEs (diarrhea, vomiting, ischemic stroke, and pneumonitis, one case each) related to ipatasertib, from which they recovered. Confirmed overall response rate was 31.1% (95% CI: 12.1-58.5%), clinical benefit rate 84.6% (95% CI: 53.7-97.3%), and median progression-free survival 16.4 months (95% CI: 9.4-NR), with 47.3% of patients being progression-free at 18 months.CONCLUSIONSThese results support ipatasertib plus HP as a safe and promising maintenance strategy for HER2-positive breast tumors harboring PIK3CAmut.CLINICALTRIALSgov registration: NCT04253561.
{"title":"Addition of ipatasertib to dual anti-HER2 maintenance therapy in HER2-positive metastatic breast tumors with PIK3CA mutations: the phase 1b SOLTI-1507 IPATHER trial.","authors":"Mafalda Oliveira,Eva Ciruelos,Guillermo Villacampa,Serafin Morales,Javier Salvador Bofill,Vanesa Quiroga,Joaquín Gavilá,Alexandra Cortegoso,Estela Vega,Fernando Henao,Sonia Servitja,Pablo Tolosa,Samyukta Chillara,Milana Bergamino,Fernando Salvador,Mariana Paes Dias,Juan M Ferrero-Cafiero,Xavier Gonzalez Farre,Tomás Pascual,Cristina Saura","doi":"10.1158/1078-0432.ccr-25-2298","DOIUrl":"https://doi.org/10.1158/1078-0432.ccr-25-2298","url":null,"abstract":"PURPOSETo evaluate the safety and feasibility of ipatasertib combined with trastuzumab and pertuzumab as maintenance therapy after first-line treatment in patients with HER2-positive metastatic breast cancer harboring PIK3CA mutations (PIK3CAmut).EXPERIMENTAL DESIGNThis prospective, multicenter, single-arm, phase 1b study evaluated the safety and preliminary efficacy of ipatasertib, an AKT inhibitor, combined with trastuzumab and pertuzumab (HP), with or without endocrine therapy as maintainance therapy, in patients with unresectable locally advanced or metastatic PIK3CAmut, HER2-positive breast cancer following first-line induction chemotherapy and HP.RESULTSSeventeen patients were enrolled, with a median follow-up of 27.7 months. During the dose selection phase, ipatasertib at 400 mg daily (21 days on, 7 days off) with standard HP was established as the recommended phase 2 dose (RP2D). This decision was based on the absence of dose-limiting toxicities in the first six patients treated at this dose during the initial 28-day cycle, which constituted the primary endpoint. Seven (41.2%) patients experienced grade 3 treatment-related adverse events (TRAEs), with diarrhea and nausea being the most frequent. Two (11.8%) reported four serious TRAEs (diarrhea, vomiting, ischemic stroke, and pneumonitis, one case each) related to ipatasertib, from which they recovered. Confirmed overall response rate was 31.1% (95% CI: 12.1-58.5%), clinical benefit rate 84.6% (95% CI: 53.7-97.3%), and median progression-free survival 16.4 months (95% CI: 9.4-NR), with 47.3% of patients being progression-free at 18 months.CONCLUSIONSThese results support ipatasertib plus HP as a safe and promising maintenance strategy for HER2-positive breast tumors harboring PIK3CAmut.CLINICALTRIALSgov registration: NCT04253561.","PeriodicalId":10279,"journal":{"name":"Clinical Cancer Research","volume":"29 1","pages":""},"PeriodicalIF":11.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145644894","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}