Pub Date : 2024-06-12DOI: 10.1186/s13058-024-01853-2
Jiaqun Du, Xiaobang Liu, Junpeng Sun, Qian Wu, Yu Hu, Huan Shi, Li Zheng, Ying Liu, Chao Wu, Yu Gao
In this study, we prepared a bionic nanosystem of trastuzumab-functionalized SK-BR-3 cell membrane hybrid liposome-coated pyrotinib (Ptb-M-Lip-Her) for the treatment of HER2-positive breast cancer. Transmission electron microscopy, dynamic light scattering, polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting were used to verify the successful preparation of Ptb-M-Lip-Her. In vitro drug release experiments proved that Ptb-M-Lip-Her had a sustained release effect. Cell uptake experiments and in vivo imaging experiments proved that Ptb-M-Lip-Her had good targeting ability to homologous tumor cells (SK-BR-3). The results of cell experiments such as MTT, flow cytometry, immunofluorescence staining and in vivo antitumor experiments showed that Ptb-M-Lip-Her could significantly promote apoptosis and inhibit the proliferation of SK-BR-3 cells. These results clearly indicated that Ptb-M-Lip-Her may be a promising biomimetic nanosystem for targeted therapy of HER2-positive breast cancer.
{"title":"Trastuzumab-functionalized bionic pyrotinib liposomes for targeted therapy of HER2-positive breast cancer.","authors":"Jiaqun Du, Xiaobang Liu, Junpeng Sun, Qian Wu, Yu Hu, Huan Shi, Li Zheng, Ying Liu, Chao Wu, Yu Gao","doi":"10.1186/s13058-024-01853-2","DOIUrl":"10.1186/s13058-024-01853-2","url":null,"abstract":"<p><p>In this study, we prepared a bionic nanosystem of trastuzumab-functionalized SK-BR-3 cell membrane hybrid liposome-coated pyrotinib (Ptb-M-Lip-Her) for the treatment of HER2-positive breast cancer. Transmission electron microscopy, dynamic light scattering, polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting were used to verify the successful preparation of Ptb-M-Lip-Her. In vitro drug release experiments proved that Ptb-M-Lip-Her had a sustained release effect. Cell uptake experiments and in vivo imaging experiments proved that Ptb-M-Lip-Her had good targeting ability to homologous tumor cells (SK-BR-3). The results of cell experiments such as MTT, flow cytometry, immunofluorescence staining and in vivo antitumor experiments showed that Ptb-M-Lip-Her could significantly promote apoptosis and inhibit the proliferation of SK-BR-3 cells. These results clearly indicated that Ptb-M-Lip-Her may be a promising biomimetic nanosystem for targeted therapy of HER2-positive breast cancer.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11167944/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141312141","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}
Background: Immunohistochemistry (IHC) and in situ hybridization (ISH) remain standard biomarkers for therapeutic decisions in human epidermal growth factor 2 (HER2)-positive breast cancers (BCs); however, they are insufficient to explain the heterogeneous anti-HER2 response.
Methods: We aimed to investigate the correlation of in situ HER2 RNA expression (isHRE), using RNAscope, with HER2 biomarkers and the impact of isHRE on the pathological complete response (pCR) rates of 278 patients with HER2 IHC/fluorescence ISH (FISH)-positive BC receiving neoadjuvant chemotherapy and anti-HER2 targeted treatment (NCTT).
Results: We validated HER2 RNAscope scoring as a semiquantitative method to determine isHRE and showed a positive correlation between RNAscope scores and pCR rates, with particularly different rates between patients with a score of 5 versus 1-4 BCs (66.7% vs. 15.9%, p < 0.0001). There were higher RNAscope scores and pCR rates in patients with HER2 IHC 3 + versus IHC 2+/FISH + BCs and HER2 RNAscope scores and pCR rates showed similar non-linear positive correlations with HER2 copy numbers and HER2/centromere 17 ratios. Moreover, in each HER2-positive IHC/FISH category, higher pCR rates were observed in patients with RNAscope scores of 5 versus 1-4 BC. Patients achieving pCR had BCs with notably higher HER2 RNAscope scores. Multivariate analysis identified HER2 RNAscope 5 as a strong pCR predictor [odds ratio = 10.865, p < 0.001]. The combined impact of multivariate analysis-defined pCR predictors demonstrated that a higher pCR rate was observed in patients with a score of 5 versus a score of 1-4 BCs regardless of the status of hormone receptor and mono-or dual anti-HER2 blockade.
Concusions: Our results demonstrated that high isHRE (RNAscope score 5) is a strong pCR predictor in patients with HER2-positive BCs receiving NCTT, highlighting the complementary role of isHRE in stratifying HER2 status in tissue. Such stratification is relevant to anti-HER2 therapeutic efficacy, particularly using the cutoff of score 1-4 versus 5.
背景:免疫组化(IHC)和原位杂交(ISH)仍然是人类表皮生长因子2(HER2)阳性乳腺癌(BCs)治疗决策的标准生物标志物;然而,它们不足以解释异质性抗HER2反应:我们的目的是利用RNAscope研究原位HER2 RNA表达(isHRE)与HER2生物标志物的相关性,以及isHRE对278例接受新辅助化疗和抗HER2靶向治疗(NCTT)的HER2 IHC/荧光ISH(FISH)阳性乳腺癌患者病理完全反应(pCR)率的影响:我们验证了将HER2 RNAscope评分作为确定isHRE的半定量方法,结果显示RNAscope评分与pCR率呈正相关,评分为5分与1-4分的BC患者的pCR率差异尤为明显(66.7% vs. 15.9%, p Concusions):我们的研究结果表明,在接受 NCTT 治疗的 HER2 阳性 BC 患者中,高 isHRE(RNAscope 评分 5 分)是预测 pCR 的有力指标,这凸显了 isHRE 在组织中 HER2 状态分层方面的补充作用。这种分层与抗 HER2 疗效有关,特别是使用 1-4 分与 5 分的分界线。
{"title":"In situ HER2 RNA expression as a predictor of pathologic complete response of HER2-positive breast cancer patients receiving neoadjuvant chemotherapy and anti-HER2 targeted treatment.","authors":"Huang-Chun Lien, Chiao Lo, Yi-Hsuang Lee, Po-Hang Lin, Ming-Yang Wang, Wen-Hung Kuo, Li-Wei Tsai, Yen-Shen Lu, Hsiang-Wei Hu, Yu-Chia Li, Chiun-Sheng Huang","doi":"10.1186/s13058-024-01852-3","DOIUrl":"10.1186/s13058-024-01852-3","url":null,"abstract":"<p><strong>Background: </strong>Immunohistochemistry (IHC) and in situ hybridization (ISH) remain standard biomarkers for therapeutic decisions in human epidermal growth factor 2 (HER2)-positive breast cancers (BCs); however, they are insufficient to explain the heterogeneous anti-HER2 response.</p><p><strong>Methods: </strong>We aimed to investigate the correlation of in situ HER2 RNA expression (isHRE), using RNAscope, with HER2 biomarkers and the impact of isHRE on the pathological complete response (pCR) rates of 278 patients with HER2 IHC/fluorescence ISH (FISH)-positive BC receiving neoadjuvant chemotherapy and anti-HER2 targeted treatment (NCTT).</p><p><strong>Results: </strong>We validated HER2 RNAscope scoring as a semiquantitative method to determine isHRE and showed a positive correlation between RNAscope scores and pCR rates, with particularly different rates between patients with a score of 5 versus 1-4 BCs (66.7% vs. 15.9%, p < 0.0001). There were higher RNAscope scores and pCR rates in patients with HER2 IHC 3 + versus IHC 2+/FISH + BCs and HER2 RNAscope scores and pCR rates showed similar non-linear positive correlations with HER2 copy numbers and HER2/centromere 17 ratios. Moreover, in each HER2-positive IHC/FISH category, higher pCR rates were observed in patients with RNAscope scores of 5 versus 1-4 BC. Patients achieving pCR had BCs with notably higher HER2 RNAscope scores. Multivariate analysis identified HER2 RNAscope 5 as a strong pCR predictor [odds ratio = 10.865, p < 0.001]. The combined impact of multivariate analysis-defined pCR predictors demonstrated that a higher pCR rate was observed in patients with a score of 5 versus a score of 1-4 BCs regardless of the status of hormone receptor and mono-or dual anti-HER2 blockade.</p><p><strong>Concusions: </strong>Our results demonstrated that high isHRE (RNAscope score 5) is a strong pCR predictor in patients with HER2-positive BCs receiving NCTT, highlighting the complementary role of isHRE in stratifying HER2 status in tissue. Such stratification is relevant to anti-HER2 therapeutic efficacy, particularly using the cutoff of score 1-4 versus 5.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11170871/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141312140","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-10DOI: 10.1186/s13058-024-01848-z
Roberto A Leon-Ferre, Kaitlyn R Whitaker, Vera J Suman, Tanya Hoskin, Karthik V Giridhar, Raymond M Moore, Ahmad Al-Jarrad, Sarah A McLaughlin, Donald W Northfelt, Katie N Hunt, Amy Lynn Conners, Ann Moyer, Jodi M Carter, Krishna Kalari, Richard Weinshilboum, Liewei Wang, James N Ingle, Keith L Knutson, Stephen M Ansell, Judy C Boughey, Matthew P Goetz, Jose C Villasboas
Background: Tumor immune infiltration and peripheral blood immune signatures have prognostic and predictive value in breast cancer. Whether distinct peripheral blood immune phenotypes are associated with response to neoadjuvant chemotherapy (NAC) remains understudied.
Methods: Peripheral blood mononuclear cells from 126 breast cancer patients enrolled in a prospective clinical trial (NCT02022202) were analyzed using Cytometry by time-of-flight with a panel of 29 immune cell surface protein markers. Kruskal-Wallis tests or Wilcoxon rank-sum tests were used to evaluate differences in immune cell subpopulations according to breast cancer subtype and response to NAC.
Results: There were 122 evaluable samples: 47 (38.5%) from patients with hormone receptor-positive, 39 (32%) triple-negative (TNBC), and 36 (29.5%) HER2-positive breast cancer. The relative abundances of pre-treatment peripheral blood T, B, myeloid, NK, and unclassified cells did not differ according to breast cancer subtype. In TNBC, higher pre-treatment myeloid cells were associated with lower pathologic complete response (pCR) rates. In hormone receptor-positive breast cancer, lower pre-treatment CD8 + naïve and CD4 + effector memory cells re-expressing CD45RA (TEMRA) T cells were associated with more extensive residual disease after NAC. In HER2 + breast cancer, the peripheral blood immune phenotype did not differ according to NAC response.
Conclusions: Pre-treatment peripheral blood immune cell populations (myeloid in TNBC; CD8 + naïve T cells and CD4 + TEMRA cells in luminal breast cancer) were associated with response to NAC in early-stage TNBC and hormone receptor-positive breast cancers, but not in HER2 + breast cancer.
Trial registration: NCT02022202 . Registered 20 December 2013.
{"title":"Pre-treatment peripheral blood immunophenotyping and response to neoadjuvant chemotherapy in operable breast cancer.","authors":"Roberto A Leon-Ferre, Kaitlyn R Whitaker, Vera J Suman, Tanya Hoskin, Karthik V Giridhar, Raymond M Moore, Ahmad Al-Jarrad, Sarah A McLaughlin, Donald W Northfelt, Katie N Hunt, Amy Lynn Conners, Ann Moyer, Jodi M Carter, Krishna Kalari, Richard Weinshilboum, Liewei Wang, James N Ingle, Keith L Knutson, Stephen M Ansell, Judy C Boughey, Matthew P Goetz, Jose C Villasboas","doi":"10.1186/s13058-024-01848-z","DOIUrl":"10.1186/s13058-024-01848-z","url":null,"abstract":"<p><strong>Background: </strong>Tumor immune infiltration and peripheral blood immune signatures have prognostic and predictive value in breast cancer. Whether distinct peripheral blood immune phenotypes are associated with response to neoadjuvant chemotherapy (NAC) remains understudied.</p><p><strong>Methods: </strong>Peripheral blood mononuclear cells from 126 breast cancer patients enrolled in a prospective clinical trial (NCT02022202) were analyzed using Cytometry by time-of-flight with a panel of 29 immune cell surface protein markers. Kruskal-Wallis tests or Wilcoxon rank-sum tests were used to evaluate differences in immune cell subpopulations according to breast cancer subtype and response to NAC.</p><p><strong>Results: </strong>There were 122 evaluable samples: 47 (38.5%) from patients with hormone receptor-positive, 39 (32%) triple-negative (TNBC), and 36 (29.5%) HER2-positive breast cancer. The relative abundances of pre-treatment peripheral blood T, B, myeloid, NK, and unclassified cells did not differ according to breast cancer subtype. In TNBC, higher pre-treatment myeloid cells were associated with lower pathologic complete response (pCR) rates. In hormone receptor-positive breast cancer, lower pre-treatment CD8 + naïve and CD4 + effector memory cells re-expressing CD45RA (T<sub>EMRA</sub>) T cells were associated with more extensive residual disease after NAC. In HER2 + breast cancer, the peripheral blood immune phenotype did not differ according to NAC response.</p><p><strong>Conclusions: </strong>Pre-treatment peripheral blood immune cell populations (myeloid in TNBC; CD8 + naïve T cells and CD4 + T<sub>EMRA</sub> cells in luminal breast cancer) were associated with response to NAC in early-stage TNBC and hormone receptor-positive breast cancers, but not in HER2 + breast cancer.</p><p><strong>Trial registration: </strong>NCT02022202 . Registered 20 December 2013.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11165781/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141301918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-07DOI: 10.1186/s13058-024-01843-4
Muriel Lainé, Marianne E Greene, Justyna D Kurleto, Grazyna Bozek, Tiffany Leng, Rosemary J Huggins, Barry S Komm, Geoffrey L Greene
Background: Breast cancers treated with aromatase inhibitors (AIs) can develop AI resistance, which is often driven by estrogen receptor-alpha (ERα/ESR1) activating mutations, as well as by ER-independent signaling pathways. The breast ER antagonist lasofoxifene, alone or combined with palbociclib, elicited antitumor activities in a xenograft model of ER + metastatic breast cancer (mBC) harboring ESR1 mutations. The current study investigated the activity of LAS in a letrozole-resistant breast tumor model that does not have ESR1 mutations.
Methods: Letrozole-resistant, MCF7 LTLT cells tagged with luciferase-GFP were injected into the mammary duct inguinal glands of NSG mice (MIND model; 6 mice/group). Mice were randomized to vehicle, lasofoxifene ± palbociclib, fulvestrant ± palbociclib, or palbociclib alone 2-3 weeks after cell injections. Tumor growth and metastases were monitored with in vivo and ex vivo luminescence imaging, terminal tumor weight measurements, and histological analysis. The experiment was repeated with the same design and 8-9 mice in each treatment group.
Results: Western blot analysis showed that the MCF7 LTLT cells had lower ERα and higher HER2 expressions compared with normal MCF7 cells. Lasofoxifene ± palbociclib, but not fulvestrant, significantly reduced primary tumor growth versus vehicle as assessed by in vivo imaging of tumors at study ends. Percent tumor area in excised mammary glands was significantly lower for lasofoxifene plus palbociclib versus vehicle. Ki67 staining showed decreased overall tumor cell proliferation with lasofoxifene ± palbociclib. The lasofoxifene + palbociclib combination was also associated with significantly fewer bone metastases compared with vehicle. Similar results were observed in the repeat experiment.
Conclusions: In a mouse model of letrozole-resistant breast cancer with no ESR1 mutations, reduced levels of ERα, and overexpression of HER2, lasofoxifene alone or combined with palbociclib inhibited primary tumor growth more effectively than fulvestrant. Lasofoxifene plus palbociclib also reduced bone metastases. These results suggest that lasofoxifene alone or combined with a CDK4/6 inhibitor may offer benefits to patients who have ER-low and HER2-positive, AI-resistant breast cancer, independent of ESR1 mutations.
{"title":"Lasofoxifene as a potential treatment for aromatase inhibitor-resistant ER-positive breast cancer.","authors":"Muriel Lainé, Marianne E Greene, Justyna D Kurleto, Grazyna Bozek, Tiffany Leng, Rosemary J Huggins, Barry S Komm, Geoffrey L Greene","doi":"10.1186/s13058-024-01843-4","DOIUrl":"10.1186/s13058-024-01843-4","url":null,"abstract":"<p><strong>Background: </strong>Breast cancers treated with aromatase inhibitors (AIs) can develop AI resistance, which is often driven by estrogen receptor-alpha (ERα/ESR1) activating mutations, as well as by ER-independent signaling pathways. The breast ER antagonist lasofoxifene, alone or combined with palbociclib, elicited antitumor activities in a xenograft model of ER + metastatic breast cancer (mBC) harboring ESR1 mutations. The current study investigated the activity of LAS in a letrozole-resistant breast tumor model that does not have ESR1 mutations.</p><p><strong>Methods: </strong>Letrozole-resistant, MCF7 LTLT cells tagged with luciferase-GFP were injected into the mammary duct inguinal glands of NSG mice (MIND model; 6 mice/group). Mice were randomized to vehicle, lasofoxifene ± palbociclib, fulvestrant ± palbociclib, or palbociclib alone 2-3 weeks after cell injections. Tumor growth and metastases were monitored with in vivo and ex vivo luminescence imaging, terminal tumor weight measurements, and histological analysis. The experiment was repeated with the same design and 8-9 mice in each treatment group.</p><p><strong>Results: </strong>Western blot analysis showed that the MCF7 LTLT cells had lower ERα and higher HER2 expressions compared with normal MCF7 cells. Lasofoxifene ± palbociclib, but not fulvestrant, significantly reduced primary tumor growth versus vehicle as assessed by in vivo imaging of tumors at study ends. Percent tumor area in excised mammary glands was significantly lower for lasofoxifene plus palbociclib versus vehicle. Ki67 staining showed decreased overall tumor cell proliferation with lasofoxifene ± palbociclib. The lasofoxifene + palbociclib combination was also associated with significantly fewer bone metastases compared with vehicle. Similar results were observed in the repeat experiment.</p><p><strong>Conclusions: </strong>In a mouse model of letrozole-resistant breast cancer with no ESR1 mutations, reduced levels of ERα, and overexpression of HER2, lasofoxifene alone or combined with palbociclib inhibited primary tumor growth more effectively than fulvestrant. Lasofoxifene plus palbociclib also reduced bone metastases. These results suggest that lasofoxifene alone or combined with a CDK4/6 inhibitor may offer benefits to patients who have ER-low and HER2-positive, AI-resistant breast cancer, independent of ESR1 mutations.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11161925/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141288857","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-07DOI: 10.1186/s13058-024-01854-1
Michael F Coleman, Eylem Kulkoyluoglu Cotul, Alexander J Pfeil, Emily N Devericks, Muhammad H Safdar, Marvis Monteiro, Hao Chen, Alyssa N Ho, Numair Attaar, Hannah M Malian, Violet A Kiesel, Alexis Ramos, Matthew Smith, Heena Panchal, Adam Mailloux, Dorothy Teegarden, Stephen D Hursting, Michael K Wendt
Background: Metabolic plasticity mediates breast cancer survival, growth, and immune evasion during metastasis. However, how tumor cell metabolism is influenced by and feeds back to regulate breast cancer progression are not fully understood. We identify hypoxia-mediated suppression of pyruvate carboxylase (PC), and subsequent induction of lactate production, as a metabolic regulator of immunosuppression.
Methods: We used qPCR, immunoblot, and reporter assays to characterize repression of PC in hypoxic primary tumors. Steady state metabolomics were used to identify changes in metabolite pools upon PC depletion. In vivo tumor growth and metastasis assays were used to evaluate the impact of PC manipulation and pharmacologic inhibition of lactate transporters. Immunohistochemistry, flow cytometry, and global gene expression analyzes of tumor tissue were employed to characterize the impact of PC depletion on tumor immunity.
Results: PC is essential for metastatic colonization of the lungs. In contrast, depletion of PC in tumor cells promotes primary tumor growth. This effect was only observed in immune competent animals, supporting the hypothesis that repression of PC can suppress anti-tumor immunity. Exploring key differences between the pulmonary and mammary environments, we demonstrate that hypoxia potently downregulated PC. In the absence of PC, tumor cells produce more lactate and undergo less oxidative phosphorylation. Inhibition of lactate metabolism was sufficient to restore T cell populations to PC-depleted mammary tumors.
Conclusions: We present a dimorphic role for PC in primary mammary tumors vs. pulmonary metastases. These findings highlight a key contextual role for PC-directed lactate production as a metabolic nexus connecting hypoxia and antitumor immunity.
背景:代谢可塑性介导乳腺癌的生存、生长和转移过程中的免疫逃避。然而,人们对肿瘤细胞的新陈代谢如何受乳腺癌进展的影响并反作用于乳腺癌进展还不完全了解。我们发现缺氧介导的丙酮酸羧化酶(PC)抑制和随后的乳酸生成诱导是免疫抑制的代谢调节因子:方法:我们使用 qPCR、免疫印迹和报告基因检测法来描述缺氧原发性肿瘤中 PC 受抑制的特征。稳态代谢组学用于确定PC耗竭时代谢物池的变化。体内肿瘤生长和转移试验用于评估 PC 操作和药物抑制乳酸转运体的影响。免疫组化、流式细胞术和肿瘤组织的全基因表达分析被用来描述PC耗竭对肿瘤免疫的影响:结果:PC对肺部转移性定植至关重要。相反,肿瘤细胞中 PC 的耗竭会促进原发性肿瘤的生长。只有在具有免疫能力的动物中才能观察到这种效应,这支持了抑制 PC 可抑制抗肿瘤免疫的假设。在探索肺部和乳腺环境的关键差异时,我们发现缺氧能有效下调 PC。在缺乏 PC 的情况下,肿瘤细胞会产生更多乳酸,氧化磷酸化过程也会减少。抑制乳酸代谢足以恢复缺失 PC 的乳腺肿瘤中的 T 细胞群:结论:我们发现 PC 在原发性乳腺肿瘤与肺转移瘤中的作用存在二态性。这些发现凸显了 PC 主导的乳酸生成作为连接缺氧和抗肿瘤免疫的代谢纽带的关键作用。
{"title":"Hypoxia-mediated repression of pyruvate carboxylase drives immunosuppression.","authors":"Michael F Coleman, Eylem Kulkoyluoglu Cotul, Alexander J Pfeil, Emily N Devericks, Muhammad H Safdar, Marvis Monteiro, Hao Chen, Alyssa N Ho, Numair Attaar, Hannah M Malian, Violet A Kiesel, Alexis Ramos, Matthew Smith, Heena Panchal, Adam Mailloux, Dorothy Teegarden, Stephen D Hursting, Michael K Wendt","doi":"10.1186/s13058-024-01854-1","DOIUrl":"10.1186/s13058-024-01854-1","url":null,"abstract":"<p><strong>Background: </strong>Metabolic plasticity mediates breast cancer survival, growth, and immune evasion during metastasis. However, how tumor cell metabolism is influenced by and feeds back to regulate breast cancer progression are not fully understood. We identify hypoxia-mediated suppression of pyruvate carboxylase (PC), and subsequent induction of lactate production, as a metabolic regulator of immunosuppression.</p><p><strong>Methods: </strong>We used qPCR, immunoblot, and reporter assays to characterize repression of PC in hypoxic primary tumors. Steady state metabolomics were used to identify changes in metabolite pools upon PC depletion. In vivo tumor growth and metastasis assays were used to evaluate the impact of PC manipulation and pharmacologic inhibition of lactate transporters. Immunohistochemistry, flow cytometry, and global gene expression analyzes of tumor tissue were employed to characterize the impact of PC depletion on tumor immunity.</p><p><strong>Results: </strong>PC is essential for metastatic colonization of the lungs. In contrast, depletion of PC in tumor cells promotes primary tumor growth. This effect was only observed in immune competent animals, supporting the hypothesis that repression of PC can suppress anti-tumor immunity. Exploring key differences between the pulmonary and mammary environments, we demonstrate that hypoxia potently downregulated PC. In the absence of PC, tumor cells produce more lactate and undergo less oxidative phosphorylation. Inhibition of lactate metabolism was sufficient to restore T cell populations to PC-depleted mammary tumors.</p><p><strong>Conclusions: </strong>We present a dimorphic role for PC in primary mammary tumors vs. pulmonary metastases. These findings highlight a key contextual role for PC-directed lactate production as a metabolic nexus connecting hypoxia and antitumor immunity.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11161980/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141288856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-06DOI: 10.1186/s13058-024-01847-0
Xuran Zhang, Ke An, Xin Ge, Yuanyuan Sun, Jingyao Wei, Weihong Ren, Han Wang, Yueqin Wang, Yue Du, Lulu He, Ouwen Li, Shaoxuan Zhou, Yong Shi, Tong Ren, Yun-Gui Yang, Quancheng Kan, Xin Tian
Background: RNA m5C methylation has been extensively implicated in the occurrence and development of tumors. As the main methyltransferase, NSUN2 plays a crucial regulatory role across diverse tumor types. However, the precise impact of NSUN2-mediated m5C modification on breast cancer (BC) remains unclear. Our study aims to elucidate the molecular mechanism underlying how NSUN2 regulates the target gene HGH1 (also known as FAM203) through m5C modification, thereby promoting BC progression. Additionally, this study targets at preliminarily clarifying the biological roles of NSUN2 and HGH1 in BC.
Methods: Tumor and adjacent tissues from 5 BC patients were collected, and the m5C modification target HGH1 in BC was screened through RNA sequencing (RNA-seq) and single-base resolution m5C methylation sequencing (RNA-BisSeq). Methylation RNA immunoprecipitation-qPCR (MeRIP-qPCR) and RNA-binding protein immunoprecipitation-qPCR (RIP-qPCR) confirmed that the methylation molecules NSUN2 and YBX1 specifically recognized and bound to HGH1 through m5C modification. In addition, proteomics, co-immunoprecipitation (co-IP), and Ribosome sequencing (Ribo-Seq) were used to explore the biological role of HGH1 in BC.
Results: As the main m5C methylation molecule, NSUN2 is abnormally overexpressed in BC and increases the overall level of RNA m5C. Knocking down NSUN2 can inhibit BC progression in vitro or in vivo. Combined RNA-seq and RNA-BisSeq analysis identified HGH1 as a potential target of abnormal m5C modifications. We clarified the mechanism by which NSUN2 regulates HGH1 expression through m5C modification, a process that involves interactions with the YBX1 protein, which collectively impacts mRNA stability and protein synthesis. Furthermore, this study is the first to reveal the binding interaction between HGH1 and the translation elongation factor EEF2, providing a comprehensive understanding of its ability to regulate transcript translation efficiency and protein synthesis in BC cells.
Conclusions: This study preliminarily clarifies the regulatory role of the NSUN2-YBX1-m5C-HGH1 axis from post-transcriptional modification to protein translation, revealing the key role of abnormal RNA m5C modification in BC and suggesting that HGH1 may be a new epigenetic biomarker and potential therapeutic target for BC.
背景:RNA m5C甲基化与肿瘤的发生和发展有着广泛的联系。作为主要的甲基转移酶,NSUN2 在多种肿瘤类型中发挥着重要的调控作用。然而,NSUN2介导的m5C修饰对乳腺癌(BC)的确切影响仍不清楚。我们的研究旨在阐明NSUN2如何通过m5C修饰调控靶基因HGH1(又称FAM203),从而促进乳腺癌进展的分子机制。此外,本研究还旨在初步阐明NSUN2和HGH1在BC中的生物学作用:方法:收集5例BC患者的肿瘤和邻近组织,通过RNA测序(RNA-seq)和单碱基分辨率m5C甲基化测序(RNA-BisSeq)筛选BC中的m5C修饰靶点HGH1。甲基化RNA免疫沉淀-qPCR(MeRIP-qPCR)和RNA结合蛋白免疫沉淀-qPCR(RIP-qPCR)证实,甲基化分子NSUN2和YBX1通过m5C修饰与HGH1特异性识别和结合。此外,蛋白质组学、共免疫沉淀(co-immunoprecipitation,co-IP)和核糖体测序(Ribo-Seq)也被用于探讨HGH1在BC中的生物学作用:结果:作为主要的m5C甲基化分子,NSUN2在BC中异常过表达,并增加了RNA m5C的整体水平。敲除 NSUN2 可抑制 BC 在体外或体内的进展。结合RNA-seq和RNA-BisSeq分析发现,HGH1是m5C异常修饰的潜在靶点。我们阐明了NSUN2通过m5C修饰调控HGH1表达的机制,这一过程涉及与YBX1蛋白的相互作用,共同影响mRNA的稳定性和蛋白质的合成。此外,该研究还首次揭示了HGH1与翻译延伸因子EEF2之间的结合相互作用,使人们对其调控BC细胞中转录本翻译效率和蛋白质合成的能力有了全面的了解:本研究初步阐明了NSUN2-YBX1-m5C-HGH1轴从转录后修饰到蛋白质翻译的调控作用,揭示了RNA m5C修饰异常在BC中的关键作用,提示HGH1可能是BC新的表观遗传学生物标志物和潜在的治疗靶点。
{"title":"NSUN2/YBX1 promotes the progression of breast cancer by enhancing HGH1 mRNA stability through m<sup>5</sup>C methylation.","authors":"Xuran Zhang, Ke An, Xin Ge, Yuanyuan Sun, Jingyao Wei, Weihong Ren, Han Wang, Yueqin Wang, Yue Du, Lulu He, Ouwen Li, Shaoxuan Zhou, Yong Shi, Tong Ren, Yun-Gui Yang, Quancheng Kan, Xin Tian","doi":"10.1186/s13058-024-01847-0","DOIUrl":"10.1186/s13058-024-01847-0","url":null,"abstract":"<p><strong>Background: </strong>RNA m<sup>5</sup>C methylation has been extensively implicated in the occurrence and development of tumors. As the main methyltransferase, NSUN2 plays a crucial regulatory role across diverse tumor types. However, the precise impact of NSUN2-mediated m<sup>5</sup>C modification on breast cancer (BC) remains unclear. Our study aims to elucidate the molecular mechanism underlying how NSUN2 regulates the target gene HGH1 (also known as FAM203) through m<sup>5</sup>C modification, thereby promoting BC progression. Additionally, this study targets at preliminarily clarifying the biological roles of NSUN2 and HGH1 in BC.</p><p><strong>Methods: </strong>Tumor and adjacent tissues from 5 BC patients were collected, and the m<sup>5</sup>C modification target HGH1 in BC was screened through RNA sequencing (RNA-seq) and single-base resolution m<sup>5</sup>C methylation sequencing (RNA-BisSeq). Methylation RNA immunoprecipitation-qPCR (MeRIP-qPCR) and RNA-binding protein immunoprecipitation-qPCR (RIP-qPCR) confirmed that the methylation molecules NSUN2 and YBX1 specifically recognized and bound to HGH1 through m<sup>5</sup>C modification. In addition, proteomics, co-immunoprecipitation (co-IP), and Ribosome sequencing (Ribo-Seq) were used to explore the biological role of HGH1 in BC.</p><p><strong>Results: </strong>As the main m<sup>5</sup>C methylation molecule, NSUN2 is abnormally overexpressed in BC and increases the overall level of RNA m<sup>5</sup>C. Knocking down NSUN2 can inhibit BC progression in vitro or in vivo. Combined RNA-seq and RNA-BisSeq analysis identified HGH1 as a potential target of abnormal m<sup>5</sup>C modifications. We clarified the mechanism by which NSUN2 regulates HGH1 expression through m<sup>5</sup>C modification, a process that involves interactions with the YBX1 protein, which collectively impacts mRNA stability and protein synthesis. Furthermore, this study is the first to reveal the binding interaction between HGH1 and the translation elongation factor EEF2, providing a comprehensive understanding of its ability to regulate transcript translation efficiency and protein synthesis in BC cells.</p><p><strong>Conclusions: </strong>This study preliminarily clarifies the regulatory role of the NSUN2-YBX1-m<sup>5</sup>C-HGH1 axis from post-transcriptional modification to protein translation, revealing the key role of abnormal RNA m<sup>5</sup>C modification in BC and suggesting that HGH1 may be a new epigenetic biomarker and potential therapeutic target for BC.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11155144/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141285174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-04DOI: 10.1186/s13058-024-01851-4
Marisa M Faraldo, Mathilde Romagnoli, Loane Wallon, Pierre Dubus, Marie-Ange Deugnier, Silvia Fre
Background: The aberrant amplification of mammary luminal progenitors is at the origin of basal-like breast cancers associated with BRCA1 mutations. Integrins mediate cell-matrix adhesion and transmit mechanical and chemical signals that drive epithelial stem cell functions and regulate tumor progression, metastatic reactivation, and resistance to targeted therapies. Consistently, we have recently shown that laminin-binding integrins are essential for the expansion and differentiation of mammary luminal progenitors in physiological conditions. As over-expression of the laminin-binding α6 integrin (Itgα6) is associated with poor prognosis and reduced survival in breast cancer, we here investigate the role of Itgα6 in mammary tumorigenesis.
Methods: We used Blg-Cre; Brca1F/F; Trp53F/F mice, a model that phenocopies human basal-like breast cancer with BRCA1 mutations. We generated mutant mice proficient or deficient in Itgα6 expression and followed tumor formation. Mammary tumors and pretumoral tissues were characterized by immunohistochemistry, flow cytometry, RT-qPCR, Western blotting and organoid cultures. Clonogenicity of luminal progenitors from preneoplastic glands was studied in 3D Matrigel cultures.
Results: We show that Itga6 deletion favors activation of p16 cell cycle inhibitor in the preneoplastic tissue. Subsequently, the amplification of luminal progenitors, the cell of origin of Brca1-deficient tumors, is restrained in Itgα6-deficient gland. In addition, the partial EMT program operating in Brca1/p53-deficient epithelium is attenuated in the absence of Itgα6. As a consequence of these events, mammary tumor formation is delayed in Itgα6-deficient mice. After tumor formation, the lack of Itgα6 does not affect tumor growth but rather alters their differentiation, resulting in reduced expression of basal cell markers.
Conclusions: Our data indicate that Itgα6 has a pro-tumorigenic role in Blg-Cre; Brca1F/F; Trp53F/F mice developing basal-like mammary tumors. In particular, we reveal that Itgα6 is required for the luminal progenitor expansion and the aberrant partial EMT program that precedes the formation of BRCA1 deficient tumors.
{"title":"Alpha-6 integrin deletion delays the formation of Brca1/p53-deficient basal-like breast tumors by restricting luminal progenitor cell expansion.","authors":"Marisa M Faraldo, Mathilde Romagnoli, Loane Wallon, Pierre Dubus, Marie-Ange Deugnier, Silvia Fre","doi":"10.1186/s13058-024-01851-4","DOIUrl":"10.1186/s13058-024-01851-4","url":null,"abstract":"<p><strong>Background: </strong>The aberrant amplification of mammary luminal progenitors is at the origin of basal-like breast cancers associated with BRCA1 mutations. Integrins mediate cell-matrix adhesion and transmit mechanical and chemical signals that drive epithelial stem cell functions and regulate tumor progression, metastatic reactivation, and resistance to targeted therapies. Consistently, we have recently shown that laminin-binding integrins are essential for the expansion and differentiation of mammary luminal progenitors in physiological conditions. As over-expression of the laminin-binding α6 integrin (Itgα6) is associated with poor prognosis and reduced survival in breast cancer, we here investigate the role of Itgα6 in mammary tumorigenesis.</p><p><strong>Methods: </strong>We used Blg-Cre; Brca1<sup>F/F</sup>; Trp53<sup>F/F</sup> mice, a model that phenocopies human basal-like breast cancer with BRCA1 mutations. We generated mutant mice proficient or deficient in Itgα6 expression and followed tumor formation. Mammary tumors and pretumoral tissues were characterized by immunohistochemistry, flow cytometry, RT-qPCR, Western blotting and organoid cultures. Clonogenicity of luminal progenitors from preneoplastic glands was studied in 3D Matrigel cultures.</p><p><strong>Results: </strong>We show that Itga6 deletion favors activation of p16 cell cycle inhibitor in the preneoplastic tissue. Subsequently, the amplification of luminal progenitors, the cell of origin of Brca1-deficient tumors, is restrained in Itgα6-deficient gland. In addition, the partial EMT program operating in Brca1/p53-deficient epithelium is attenuated in the absence of Itgα6. As a consequence of these events, mammary tumor formation is delayed in Itgα6-deficient mice. After tumor formation, the lack of Itgα6 does not affect tumor growth but rather alters their differentiation, resulting in reduced expression of basal cell markers.</p><p><strong>Conclusions: </strong>Our data indicate that Itgα6 has a pro-tumorigenic role in Blg-Cre; Brca1<sup>F/F</sup>; Trp53<sup>F/F</sup> mice developing basal-like mammary tumors. In particular, we reveal that Itgα6 is required for the luminal progenitor expansion and the aberrant partial EMT program that precedes the formation of BRCA1 deficient tumors.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11151721/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141248935","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-03DOI: 10.1186/s13058-024-01837-2
Pere Miquel Morla-Barcelo, David Laguna-Macarrilla, Octavi Cordoba, Gabriel Matheu, Jordi Oliver, Pilar Roca, Mercedes Nadal-Serrano, Jorge Sastre-Serra
Background: Early-stage invasive ductal carcinoma displays high survival rates due to early detection and treatments. However, there is still a chance of relapse of 3-15% after treatment. The aim of this study was to uncover the distinctive transcriptomic characteristics and monitoring prognosis potential of peritumoral tissue in early-stage cases.
Methods: RNA was isolated from tumoral, peritumoral, and non-tumoral breast tissue from surgical resection of 10 luminal early-stage invasive ductal carcinoma patients. Transcriptome expression profiling for differentially expressed genes (DEGs) identification was carried out through microarray analysis. Gene Ontology and KEGG pathways enrichment analysis were explored for functional characterization of identified DEGs. Protein-Protein Interactions (PPI) networks analysis was performed to identify hub nodes of peritumoral tissue alterations and correlated with Overall Survival and Relapse Free Survival.
Results: DEGs closely related with cell migration, extracellular matrix organization, and cell cycle were upregulated in peritumoral tissue compared to non-tumoral. Analyzing PPI networks, we observed that the proximity to tumor leads to the alteration of gene modules involved in cell proliferation and differentiation signaling pathways. In fact, in the peritumoral area were identified the top ten upregulated hub nodes including CDK1, ESR1, NOP58, PCNA, EZH2, PPP1CA, BUB1, TGFBR1, CXCR4, and CCND1. A signature performed by four of these hub nodes (CDK1, PCNA, EZH2, and BUB1) was associated with relapse events in untreated luminal breast cancer patients.
Conclusions: In conclusion, our study characterizes in depth breast peritumoral tissue providing clues on the changes that tumor signaling could cause in patients with early-stage breast cancer. We propose that the use of a four gene signature could help to predict local relapse. Overall, our results highlight the value of peritumoral tissue as a potential source of new biomarkers for early detection of relapse and improvement in invasive ductal carcinoma patient's prognosis.
{"title":"Unraveling malignant phenotype of peritumoral tissue: transcriptomic insights into early-stage breast cancer.","authors":"Pere Miquel Morla-Barcelo, David Laguna-Macarrilla, Octavi Cordoba, Gabriel Matheu, Jordi Oliver, Pilar Roca, Mercedes Nadal-Serrano, Jorge Sastre-Serra","doi":"10.1186/s13058-024-01837-2","DOIUrl":"10.1186/s13058-024-01837-2","url":null,"abstract":"<p><strong>Background: </strong>Early-stage invasive ductal carcinoma displays high survival rates due to early detection and treatments. However, there is still a chance of relapse of 3-15% after treatment. The aim of this study was to uncover the distinctive transcriptomic characteristics and monitoring prognosis potential of peritumoral tissue in early-stage cases.</p><p><strong>Methods: </strong>RNA was isolated from tumoral, peritumoral, and non-tumoral breast tissue from surgical resection of 10 luminal early-stage invasive ductal carcinoma patients. Transcriptome expression profiling for differentially expressed genes (DEGs) identification was carried out through microarray analysis. Gene Ontology and KEGG pathways enrichment analysis were explored for functional characterization of identified DEGs. Protein-Protein Interactions (PPI) networks analysis was performed to identify hub nodes of peritumoral tissue alterations and correlated with Overall Survival and Relapse Free Survival.</p><p><strong>Results: </strong>DEGs closely related with cell migration, extracellular matrix organization, and cell cycle were upregulated in peritumoral tissue compared to non-tumoral. Analyzing PPI networks, we observed that the proximity to tumor leads to the alteration of gene modules involved in cell proliferation and differentiation signaling pathways. In fact, in the peritumoral area were identified the top ten upregulated hub nodes including CDK1, ESR1, NOP58, PCNA, EZH2, PPP1CA, BUB1, TGFBR1, CXCR4, and CCND1. A signature performed by four of these hub nodes (CDK1, PCNA, EZH2, and BUB1) was associated with relapse events in untreated luminal breast cancer patients.</p><p><strong>Conclusions: </strong>In conclusion, our study characterizes in depth breast peritumoral tissue providing clues on the changes that tumor signaling could cause in patients with early-stage breast cancer. We propose that the use of a four gene signature could help to predict local relapse. Overall, our results highlight the value of peritumoral tissue as a potential source of new biomarkers for early detection of relapse and improvement in invasive ductal carcinoma patient's prognosis.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11145834/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141238619","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-06-03DOI: 10.1186/s13058-024-01840-7
Constance Boissin, Yinxi Wang, Abhinav Sharma, Philippe Weitz, Emelie Karlsson, Stephanie Robertson, Johan Hartman, Mattias Rantalainen
Background: Nottingham histological grade (NHG) is a well established prognostic factor in breast cancer histopathology but has a high inter-assessor variability with many tumours being classified as intermediate grade, NHG2. Here, we evaluate if DeepGrade, a previously developed model for risk stratification of resected tumour specimens, could be applied to risk-stratify tumour biopsy specimens.
Methods: A total of 11,955,755 tiles from 1169 whole slide images of preoperative biopsies from 896 patients diagnosed with breast cancer in Stockholm, Sweden, were included. DeepGrade, a deep convolutional neural network model, was applied for the prediction of low- and high-risk tumours. It was evaluated against clinically assigned grades NHG1 and NHG3 on the biopsy specimen but also against the grades assigned to the corresponding resection specimen using area under the operating curve (AUC). The prognostic value of the DeepGrade model in the biopsy setting was evaluated using time-to-event analysis.
Results: Based on preoperative biopsy images, the DeepGrade model predicted resected tumour cases of clinical grades NHG1 and NHG3 with an AUC of 0.908 (95% CI: 0.88; 0.93). Furthermore, out of the 432 resected clinically-assigned NHG2 tumours, 281 (65%) were classified as DeepGrade-low and 151 (35%) as DeepGrade-high. Using a multivariable Cox proportional hazards model the hazard ratio between DeepGrade low- and high-risk groups was estimated as 2.01 (95% CI: 1.06; 3.79).
Conclusions: DeepGrade provided prediction of tumour grades NHG1 and NHG3 on the resection specimen using only the biopsy specimen. The results demonstrate that the DeepGrade model can provide decision support to identify high-risk tumours based on preoperative biopsies, thus improving early treatment decisions.
{"title":"Deep learning-based risk stratification of preoperative breast biopsies using digital whole slide images.","authors":"Constance Boissin, Yinxi Wang, Abhinav Sharma, Philippe Weitz, Emelie Karlsson, Stephanie Robertson, Johan Hartman, Mattias Rantalainen","doi":"10.1186/s13058-024-01840-7","DOIUrl":"10.1186/s13058-024-01840-7","url":null,"abstract":"<p><strong>Background: </strong>Nottingham histological grade (NHG) is a well established prognostic factor in breast cancer histopathology but has a high inter-assessor variability with many tumours being classified as intermediate grade, NHG2. Here, we evaluate if DeepGrade, a previously developed model for risk stratification of resected tumour specimens, could be applied to risk-stratify tumour biopsy specimens.</p><p><strong>Methods: </strong>A total of 11,955,755 tiles from 1169 whole slide images of preoperative biopsies from 896 patients diagnosed with breast cancer in Stockholm, Sweden, were included. DeepGrade, a deep convolutional neural network model, was applied for the prediction of low- and high-risk tumours. It was evaluated against clinically assigned grades NHG1 and NHG3 on the biopsy specimen but also against the grades assigned to the corresponding resection specimen using area under the operating curve (AUC). The prognostic value of the DeepGrade model in the biopsy setting was evaluated using time-to-event analysis.</p><p><strong>Results: </strong>Based on preoperative biopsy images, the DeepGrade model predicted resected tumour cases of clinical grades NHG1 and NHG3 with an AUC of 0.908 (95% CI: 0.88; 0.93). Furthermore, out of the 432 resected clinically-assigned NHG2 tumours, 281 (65%) were classified as DeepGrade-low and 151 (35%) as DeepGrade-high. Using a multivariable Cox proportional hazards model the hazard ratio between DeepGrade low- and high-risk groups was estimated as 2.01 (95% CI: 1.06; 3.79).</p><p><strong>Conclusions: </strong>DeepGrade provided prediction of tumour grades NHG1 and NHG3 on the resection specimen using only the biopsy specimen. The results demonstrate that the DeepGrade model can provide decision support to identify high-risk tumours based on preoperative biopsies, thus improving early treatment decisions.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11145850/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141238617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-05-31DOI: 10.1186/s13058-024-01834-5
Esther M John, Jocelyn Koo, Amanda I Phipps, Teri A Longacre, Allison W Kurian, Sue A Ingles, Anna H Wu, Lisa M Hines
Background: Associations between reproductive factors and risk of breast cancer differ by subtype defined by joint estrogen receptor (ER), progesterone receptor (PR), and HER2 expression status. Racial and ethnic differences in the incidence of breast cancer subtypes suggest etiologic heterogeneity, yet data are limited because most studies have included non-Hispanic White women only.
Methods: We analyzed harmonized data for 2,794 breast cancer cases and 4,579 controls, of whom 90% self-identified as African American, Asian American or Hispanic. Questionnaire data were pooled from three population-based studies conducted in California and data on tumor characteristics were obtained from the California Cancer Registry. The study sample included 1,530 luminal A (ER-positive and/or PR-positive, HER2-negative), 442 luminal B (ER-positive and/or PR-positive, HER2-positive), 578 triple-negative (TN; ER-negative, PR-negative, HER2-negative), and 244 HER2-enriched (ER-negative, PR-negative, HER2-positive) cases. We used multivariable unconditional logistic regression models to estimate subtype-specific ORs and 95% confidence intervals associated with parity, breast-feeding, and other reproductive characteristics by menopausal status and race and ethnicity.
Results: Subtype-specific associations with reproductive factors revealed some notable differences by menopausal status and race and ethnicity. Specifically, higher parity without breast-feeding was associated with higher risk of luminal A and TN subtypes among premenopausal African American women. In contrast, among Asian American and Hispanic women, regardless of menopausal status, higher parity with a breast-feeding history was associated with lower risk of luminal A subtype. Among premenopausal women only, luminal A subtype was associated with older age at first full-term pregnancy (FTP), longer interval between menarche and first FTP, and shorter interval since last FTP, with similar OR estimates across the three racial and ethnic groups.
Conclusions: Subtype-specific associations with reproductive factors overall and by menopausal status, and race and ethnicity, showed some differences, underscoring that understanding etiologic heterogeneity in racially and ethnically diverse study samples is essential. Breast-feeding is likely the only reproductive factor that is potentially modifiable. Targeted efforts to promote and facilitate breast-feeding could help mitigate the adverse effects of higher parity among premenopausal African American women.
背景:根据雌激素受体(ER)、孕激素受体(PR)和 HER2 联合表达状态所定义的亚型,生殖因素与乳腺癌风险之间的关系有所不同。乳腺癌亚型发病率的种族和民族差异表明病因存在异质性,但由于大多数研究仅包括非西班牙裔白人妇女,因此数据有限:我们分析了 2,794 例乳腺癌病例和 4,579 例对照的统一数据,其中 90% 的病例自认为是非裔美国人、亚裔美国人或西班牙裔美国人。问卷数据来自加利福尼亚州进行的三项人群研究,肿瘤特征数据来自加利福尼亚州癌症登记处。研究样本包括 1,530 例管腔 A 型(ER 阳性和/或 PR 阳性、HER2 阴性)、442 例管腔 B 型(ER 阳性和/或 PR 阳性、HER2 阳性)、578 例三联阴性(TN;ER 阴性、PR 阴性、HER2 阴性)和 244 例 HER2 富集型(ER 阴性、PR 阴性、HER2 阳性)病例。我们使用多变量无条件逻辑回归模型估算了亚型特异性 ORs 和 95% 的置信区间,这些 ORs 与更年期状态、种族和民族的奇偶数、母乳喂养和其他生殖特征有关:亚型特异性与生殖因素的关系显示出绝经状态、种族和民族的一些显著差异。具体而言,在绝经前的非裔美国妇女中,较高的奇偶数(无母乳喂养)与较高的管腔 A 和 TN 亚型风险相关。相比之下,在亚裔美国人和西班牙裔妇女中,无论绝经状态如何,有哺乳史的高奇偶性与管腔 A 亚型的低风险相关。仅在绝经前妇女中,管腔 A 亚型与首次足月妊娠(FTP)年龄较大、月经初潮与首次足月妊娠之间的间隔时间较长以及距上次足月妊娠的间隔时间较短有关,三个种族和族裔群体的OR估计值相似:总的来说,亚型与生殖因素的特异性关联以及绝经状态、种族和民族的特异性关联显示出一些差异,这突出表明了解种族和民族多样性研究样本中的病因异质性至关重要。母乳喂养可能是唯一有可能改变的生殖因素。有针对性地推广和促进母乳喂养有助于减轻绝经前非洲裔美国妇女中较高的奇偶性所带来的不利影响。
{"title":"Reproductive characteristics, menopausal status, race and ethnicity, and risk of breast cancer subtypes defined by ER, PR and HER2 status: the Breast Cancer Etiology in Minorities study.","authors":"Esther M John, Jocelyn Koo, Amanda I Phipps, Teri A Longacre, Allison W Kurian, Sue A Ingles, Anna H Wu, Lisa M Hines","doi":"10.1186/s13058-024-01834-5","DOIUrl":"10.1186/s13058-024-01834-5","url":null,"abstract":"<p><strong>Background: </strong>Associations between reproductive factors and risk of breast cancer differ by subtype defined by joint estrogen receptor (ER), progesterone receptor (PR), and HER2 expression status. Racial and ethnic differences in the incidence of breast cancer subtypes suggest etiologic heterogeneity, yet data are limited because most studies have included non-Hispanic White women only.</p><p><strong>Methods: </strong>We analyzed harmonized data for 2,794 breast cancer cases and 4,579 controls, of whom 90% self-identified as African American, Asian American or Hispanic. Questionnaire data were pooled from three population-based studies conducted in California and data on tumor characteristics were obtained from the California Cancer Registry. The study sample included 1,530 luminal A (ER-positive and/or PR-positive, HER2-negative), 442 luminal B (ER-positive and/or PR-positive, HER2-positive), 578 triple-negative (TN; ER-negative, PR-negative, HER2-negative), and 244 HER2-enriched (ER-negative, PR-negative, HER2-positive) cases. We used multivariable unconditional logistic regression models to estimate subtype-specific ORs and 95% confidence intervals associated with parity, breast-feeding, and other reproductive characteristics by menopausal status and race and ethnicity.</p><p><strong>Results: </strong>Subtype-specific associations with reproductive factors revealed some notable differences by menopausal status and race and ethnicity. Specifically, higher parity without breast-feeding was associated with higher risk of luminal A and TN subtypes among premenopausal African American women. In contrast, among Asian American and Hispanic women, regardless of menopausal status, higher parity with a breast-feeding history was associated with lower risk of luminal A subtype. Among premenopausal women only, luminal A subtype was associated with older age at first full-term pregnancy (FTP), longer interval between menarche and first FTP, and shorter interval since last FTP, with similar OR estimates across the three racial and ethnic groups.</p><p><strong>Conclusions: </strong>Subtype-specific associations with reproductive factors overall and by menopausal status, and race and ethnicity, showed some differences, underscoring that understanding etiologic heterogeneity in racially and ethnically diverse study samples is essential. Breast-feeding is likely the only reproductive factor that is potentially modifiable. Targeted efforts to promote and facilitate breast-feeding could help mitigate the adverse effects of higher parity among premenopausal African American women.</p>","PeriodicalId":49227,"journal":{"name":"Breast Cancer Research","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11143591/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141184630","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}