Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.458
Filtrer
1.
Zhonghua Bing Li Xue Za Zhi ; 53(7): 678-684, 2024 Jul 08.
Article de Chinois | MEDLINE | ID: mdl-38955698

RÉSUMÉ

Objective: To investigate the relationship between 21-gene recurrence risk score (21-Gene RS) and the prognosis and clinicopathological features of hormone receptor (HR) positive, HER2-negative early breast cancer patients who did not receive neoadjuvant therapy. Methods: A total of 469 patients with HR positive and HER2-negative early breast cancer who received surgical treatment in the First Affiliated Hospital, Zhejiang University School of Medicine from January 2014 to October 2017 were selected. Their clinicopathological data were retrospectively analyzed. Tumor tissue samples were collected from patients, and the expression of 21-gene was detected by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR). The 21-Gene RS was calculated according to the Trial Assigning Individualized Options for Treatment (TAILORx) RS grouping and National Surgical Adjuvant Breast and Bowel Project B-20 (NSABP B-20) RS grouping principles. Patients were divided into low (21-Gene RS<11 or 21-Gene RS<18), intermediate (11≤21-Gene RS<26 or 18≤21-Gene RS<31) and high (21-Gene RS≥26 or 21-Gene RS≥31) risk groups, and the clinicopathological features and prognostic differences of patients in different risk groups were compared. Statistical data were compared by chi-square test. Survival analysis was performed using Kaplan-Meier curve analysis and the differences between groups were compared using Log-rank test. Multivariate analysis was conducted by COX regression analysis. Results: Based on TAILORx RS grouping, the proportions of low-risk, intermediate-risk and high-risk groups among the 469 patients were 18.8% (88/469), 48.2% (226/469) and 33.0% (155/469), respectively. Based on NSABP B-20 RS grouping, the proportion of low-risk, intermediate-risk and high-risk groups were 43.1% (202/469), 37.5% (176/469) and 19.4% (91/469), respectively. The association of 21-Gene RS with histological grading, luminal typing, Ki-67 expression, and chemotherapy and treatment modalities were statistically significant (P<0.05) regardless of TAILORx RS grouping or NSABP B-20 RS grouping. Kaplan-Meier survival curve suggested poor prognosis in high-risk group (P<0.05, Log-rank test). Multivariate COX regression analysis showed that surgical method and 21-Gene RS were risk factors affecting the prognosis of patients. Conclusions: 21-Gene RS is significantly associated with the prognosis of patients with HR-positive, HER2-negative, early-stage breast cancer not receiving neoadjuvant therapy, as well as with their clinicopathological characteristics such as patients' histologic grade, luminal typing, Ki-67 expression, and whether or not they are treated with chemotherapy or other treatment modalities.The 21-Gene RS threshold of 11 and 26 or 18 and 31 can be used to grade the prognosis in Chinese patients with early-stage breast cancer. More researches are needed to guide the selection of postoperative adjuvant therapy for patients with HR-positive and HER2-negative early-stage breast cancer.


Sujet(s)
Tumeurs du sein , Récidive tumorale locale , Récepteur ErbB-2 , Humains , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Récidive tumorale locale/génétique , Pronostic , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Études rétrospectives , Récepteurs des oestrogènes/métabolisme , Récepteurs à la progestérone/métabolisme , Récepteurs à la progestérone/génétique , Adulte d'âge moyen , Facteurs de risque
2.
Front Immunol ; 15: 1349033, 2024.
Article de Anglais | MEDLINE | ID: mdl-38989283

RÉSUMÉ

Background: Extramammary Paget's disease (EMPD) is a rare epithelial malignancy, and approximately 30%-40% of EMPD patients overexpress human epidermal growth factor receptor 2 (Her-2). Currently, there are no established standard treatments for advanced EMPD while anti-Her-2 therapy is recommended for Her-2-positive cases. Case presentation: Here, we report a 51-year-old male diagnosed with advanced Her-2-positive EMPD, presenting with numerous lymph node metastases. This patient received disitamab vedotin (an antibody-drug conjugate, targeting Her-2) combined with serplulimab as first-line treatment. After seven cycles of combination therapy, the patient tolerated the treatment well and the lymph node lesions continued to shrink. However, the patient developed immunotherapy-related pneumonia following the eighth treatment. Hormone therapy was administered while all the anti-tumor therapies were halted. After the pneumonia improved, the patient underwent positron emission tomography-computed tomography, revealing a complete response to his tumor. To consolidate the effect, he received another five cycles of disitamab vedotin monotherapy as maintenance therapy, without experiencing any adverse events. To date, the patient has remained in good health without any recurrence 10 months after drug discontinuance. Conclusion: Disitamab vedotin combined with immunotherapy demonstrated a long-term clinical benefit in advanced Her-2-positive EMPD. For rare solid tumors with Her-2 overexpression, disitamab vedotin combined with immunotherapy might offer a viable therapeutic choice.


Sujet(s)
Maladie de Paget extramammaire , Récepteur ErbB-2 , Humains , Mâle , Adulte d'âge moyen , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Maladie de Paget extramammaire/traitement médicamenteux , Maladie de Paget extramammaire/thérapie , Scrotum/anatomopathologie , Résultat thérapeutique , Immunothérapie/méthodes , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Immunoconjugués/usage thérapeutique
3.
J Cancer Res Ther ; 20(3): 984-992, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-39023607

RÉSUMÉ

AIM: Although people with HER2-positive breast cancer benefit from approved HER2-targeted therapy, acquiring resistance to the therapies occurs. Animal models can play a part in gaining a deep understanding of such a process and addressing questions concerning developing and improving immunotherapy approaches. MATERIALS AND METHODS: To develop such a model, we transfected murine 4T1 cells with the pCMV6-Neo-HER2 construct and evaluated HER2 expression and its effects on the established cell line behavior in vitro and in vivo. RESULTS: Data illustrated that human HER2 protein was expressed on isolated 4T1-HER2 clones in vitro and in vivo. Except for proliferation over 48 hours, such expression did not change 4T1-HER2 characteristics compared to 4T1 in vitro. Notwithstanding the reduction in proliferation, the rate of tumorigenicity was 90% in challenged mice and Herceptin therapy significantly decreased tumors' growth and metastasis compared to the control group. CONCLUSION: We describe a murine model for HER2-positive breast cancer not only helping shed light on the mechanisms by which the tumor evades antitumor immunity but also playing a key role in making breast cancer more sensitive to novel immunotherapy modalities.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Modèles animaux de maladie humaine , Récepteur ErbB-2 , Animaux , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Souris , Femelle , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/thérapie , Lignée cellulaire tumorale , Trastuzumab/pharmacologie , Trastuzumab/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique
4.
Signal Transduct Target Ther ; 9(1): 182, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39004647

RÉSUMÉ

A dose-escalation and expansion, phase 1/2 study (ClinicalTrials.gov, NCT04818333) was conducted to assess the novel antibody-drug conjugate SHR-A1811 in pretreated HER2-altered advanced non-small cell lung cancer (NSCLC). Here, we report results from the phase 1 portion. Patients who had previously failed or were intolerant to platinum-based chemotherapy were enrolled and received SHR-A1811 intravenously at doses of 3.2 to 8.0 mg/kg every 3 weeks. Dose escalation followed a Bayesian logistic regression model that included overdose control, with subsequent selection of tolerable levels for dose expansion. Overall, 63 patients were enrolled, including 43 receiving a recommended dose for expansion of 4.8 mg/kg. All patients had HER2-mutant disease. Dose-limiting toxicity occurred in one patient in the 8.0 mg/kg dose cohort. Grade ≥ 3 treatment-related adverse events occurred in 29 (46.0%) patients. One patient in the 6.4 mg/kg cohort died due to interstitial lung disease. As of April 11, 2023, the 4.8 mg/kg cohort showed an objective response rate of 41.9% (95% CI 27.0-57.9), and a disease control rate of 95.3% (95% CI 84.2-99.4). The median duration of response was 13.7 months, with 13 of 18 responses ongoing. The median progression-free survival was 8.4 months (95% CI 7.1-15.0). SHR-A1811 demonstrated favourable safety and clinically meaningful efficacy in pretreated advanced HER2-mutant NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Immunoconjugués , Tumeurs du poumon , Mutation , Récepteur ErbB-2 , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Femelle , Adulte d'âge moyen , Mâle , Sujet âgé , Récepteur ErbB-2/génétique , Récepteur ErbB-2/antagonistes et inhibiteurs , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Adulte , Immunoconjugués/effets indésirables , Immunoconjugués/usage thérapeutique , Sujet âgé de 80 ans ou plus
5.
Wiad Lek ; 77(5): 1056-1062, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008597

RÉSUMÉ

OBJECTIVE: Aim: To clarify the association between response to Trastuzumab and molecular expression of TIM-3 and FOXP-3 immune checkpoints. PATIENTS AND METHODS: Materials and Methods: FOXP-3 and TIM-3 expression in peripheral blood was analyzed using qPCR, and the serum level of Trastuzumab was estimated using an immune sorbent enzyme assay. RESULTS: Results: During treatment with Trastuzumab, the FOXP-3 gene expression showed a significant decline throughout one year of treatment, going from 0.85 at cycle 9 to 0.75 at cycle 17. While the TIM-3 gene expression showed a significant up regulation at cycle 9 to 2.8 fold, followed by a reduction in the fold change from 2.8 to 1.7 in the font of reference gene expression. CONCLUSION: Conclusions:FOXP-3 and TIM-3 have the potential to be suggestive markers that can anticipate the response to Trastuzumab, but they are not capable of predicting the likelihood of recurrence.


Sujet(s)
Tumeurs du sein , Récepteur ErbB-2 , Trastuzumab , Humains , Trastuzumab/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Femelle , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Récepteur cellulaire-2 du virus de l'hépatite A/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Antinéoplasiques immunologiques/usage thérapeutique , Adulte d'âge moyen , Adulte , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/sang , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
6.
BMC Cancer ; 24(1): 842, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009968

RÉSUMÉ

BACKGROUND: Targeted therapy is now the standard of care in driver-oncogene-positive non-small cell lung cancer (NSCLC). Its initial clinical effects are remarkable. However, almost all patients experience treatment resistance to targeted therapy. Hence, chemotherapy is considered a subsequent treatment option. In patients with driver-oncogene-negative NSCLC, combined immune checkpoint inhibitors (ICIs) and chemotherapy as the first-line therapy has been found to be beneficial. However, the efficacy of ICI plus chemotherapy against driver-oncogene-positive NSCLC other than epidermal growth factor receptor mutation and anaplastic lymphoma kinase fusion is unclear. METHODS: Using the hospital medical records, we retrospectively reviewed advanced or recurrent NSCLC patients who were treated with chemotherapy with or without ICIs at Aichi Cancer Center Hospital between January 2014 and January 2023. Patients with druggable rare mutations such as KRAS-G12C, MET exon 14 skipping, HER2 20 insertion, BRAF-V600E mutations, and ROS1 and RET rearrangements were analyzed. RESULTS: In total, 61 patients were included in this analysis. ICI plus chemotherapy was administered in 36 patients (the ICI-chemo group) and chemotherapy in 25 patients (the chemo group). The median progression-free survival (PFS) rates were 14.0 months in the ICI-chemo group and 4.8 months in the chemo group (hazard ratio [HR] = 0.54, 95% confidence interval [CI] = 0.28-1.01). The median overall survival rates were 31.3 and 21.7 months in the ICI-chemo and chemo groups, respectively (HR = 0.70, 95% CI = 0.33-1.50). Multivariate Cox regression analysis of PFS revealed that HER2 exon 20 insertion mutation was significantly associated with a poorer PFS (HR: 2.39, 95% CI: 1.19-4.77, P = 0.014). Further, ICI-chemo treatment was significantly associated with a better PFS (HR: 0.48, 95% CI: 0.25-0.91, P = 0.025). CONCLUSION: ICI plus chemotherapy improves treatment efficacy in rare driver-oncogene-positive NSCLC.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Mutation , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome pulmonaire non à petites cellules/anatomopathologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mâle , Femelle , Études rétrospectives , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Adulte d'âge moyen , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte , Sujet âgé de 80 ans ou plus , Protéines proto-oncogènes B-raf/génétique , Récepteur ErbB-2/génétique , Protéines proto-oncogènes c-ret/génétique , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes/génétique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/génétique , Survie sans progression , Résultat thérapeutique
7.
Int J Biol Sci ; 20(9): 3497-3514, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993569

RÉSUMÉ

Resistance to HER2-targeted therapy is the major cause of treatment failure in patients with HER2+ breast cancer (BC). Given the key role of immune microenvironment in tumor development, there is a lack of an ideal prognostic model that fully accounts for immune infiltration. In this study, WGCNA analysis was performed to discover the relationship between immune-related signaling and prognosis of HER2+ BC. After Herceptin-resistant BC cell lines established, transcriptional profiles of resistant cell line and RNA-sequencing data from GSE76360 cohort were analyzed for candidate genes. 85 samples of HER2+ BC from TCGA database were analyzed by the Cox regression, XGBoost and Lasso algorithm to generalize a credible immune-related prognostic index (IRPI). Correlations between the IRPI signature and tumor microenvironment were further analyzed by multiple algorithms, including single-cell RNA sequencing data analysis. Patients with high IRPI had suppressive tumor immune microenvironment and worse prognosis. The suppression of type I interferon signaling indicated by the IRPI in Herceptin-resistant HER2+ BC was validated. And we elucidated that the suppression of cGAS-STING pathway is the key determinant underlying immune escape in Herceptin-resistant BC with high IRPI. A combination of STING agonist and DS-8201 could serve as a new strategy for Herceptin-resistant HER2+ BC.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Protéines membranaires , Nucleotidyltransferases , Récepteur ErbB-2 , Trastuzumab , Microenvironnement tumoral , Humains , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/immunologie , Femelle , Trastuzumab/usage thérapeutique , Trastuzumab/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Transduction du signal , Lignée cellulaire tumorale , Pronostic , Régulation de l'expression des gènes tumoraux
8.
Sci Rep ; 14(1): 16030, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992220

RÉSUMÉ

This study examines the biological effects of palbociclib and ribociclib in hormone receptor-positive breast cancer, pivotal to the HARMONIA prospective phase III clinical trial. We explore the downstream impacts of these CDK4/6 inhibitors, focusing on cell lines and patient-derived tumor samples. We treated HR+ breast cancer cell lines (T47D, MCF7, and BT474) with palbociclib or ribociclib (100 nM or 500 nM), alone or combined with fulvestrant (1 nM), over periods of 24, 72, or 144 h. Our assessments included PAM50 gene expression, RB1 phosphorylation, Lamin-B1 protein levels, and senescence-associated ß-galactosidase activity. We further analyzed PAM50 gene signatures from the CORALLEEN and NeoPalAna phase II trials. Both CDK4/6 inhibitors similarly inhibited proliferation across the cell lines. At 100 nM, both drugs partially reduced p-RB1, with further decreases at 500 nM over 144 h. Treatment led to reduced Lamin-B1 expression and increased senescence-associated ß-galactosidase activity. Both drugs enhanced Luminal A and reduced Luminal B and proliferation signatures at both doses. However, the HER2-enriched signature significantly diminished only at the higher dose of 500 nM. Corresponding changes were observed in tumor samples from the CORALLEEN and NeoPalAna studies. At 2 weeks of treatment, both drugs significantly reduced the HER2-enriched signature, but at surgery, this reduction was consistent only with ribociclib. Our findings suggest that while both CDK4/6 inhibitors effectively modulate key biological pathways in HR+/HER2- breast cancer, nuances in their impact, particularly on the HER2-enriched signature, are dose-dependent, influenced by the addition of fulvestrant and warrant further investigation.


Sujet(s)
Aminopyridines , Tumeurs du sein , Prolifération cellulaire , Pipérazines , Purines , Pyridines , Humains , Aminopyridines/pharmacologie , Pipérazines/pharmacologie , Purines/pharmacologie , Pyridines/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteurs des oestrogènes/métabolisme , Fulvestrant/pharmacologie , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Kinase-4 cycline-dépendante/métabolisme , Récepteurs à la progestérone/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Kinase-6 cycline-dépendante/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
9.
Breast Cancer Res ; 26(1): 107, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951909

RÉSUMÉ

PURPOSE: HER3, a member of the EGFR receptor family, plays a central role in driving oncogenic cell proliferation in breast cancer. Novel HER3 therapeutics are showing promising results while recently developed HER3 PET imaging modalities aid in predicting and assessing early treatment response. However, baseline HER3 expression, as well as changes in expression while on neoadjuvant therapy, have not been well-characterized. We conducted a prospective clinical study, pre- and post-neoadjuvant/systemic therapy, in patients with newly diagnosed breast cancer to determine HER3 expression, and to identify possible resistance mechanisms maintained through the HER3 receptor. EXPERIMENTAL DESIGN: The study was conducted between May 25, 2018 and October 12, 2019. Thirty-four patients with newly diagnosed breast cancer of any subtype (ER ± , PR ± , HER2 ±) were enrolled in the study. Two core biopsy specimens were obtained from each patient at the time of diagnosis. Four patients underwent a second research biopsy following initiation of neoadjuvant/systemic therapy or systemic therapy which we define as neoadjuvant therapy. Molecular characterization of HER3 and downstream signaling nodes of the PI3K/AKT and MAPK pathways pre- and post-initiation of therapy was performed. Transcriptional validation of finings was performed in an external dataset (GSE122630). RESULTS: Variable baseline HER3 expression was found in newly diagnosed breast cancer and correlated positively with pAKT across subtypes (r = 0.45). In patients receiving neoadjuvant/systemic therapy, changes in HER3 expression were variable. In a hormone receptor-positive (ER +/PR +/HER2-) patient, there was a statistically significant increase in HER3 expression post neoadjuvant therapy, while there was no significant change in HER3 expression in a ER +/PR +/HER2+ patient. However, both of these patients showed increased downstream signaling in the PI3K/AKT pathway. One subject with ER +/PR -/HER2- breast cancer and another subject with ER +/PR +/HER2 + breast cancer showed decreased HER3 expression. Transcriptomic findings, revealed an immune suppressive environment in patients with decreased HER3 expression post therapy. CONCLUSION: This study demonstrates variable HER3 expression across breast cancer subtypes. HER3 expression can be assessed early, post-neoadjuvant therapy, providing valuable insight into cancer biology and potentially serving as a prognostic biomarker. Clinical translation of neoadjuvant therapy assessment can be achieved using HER3 PET imaging, offering real-time information on tumor biology and guiding personalized treatment for breast cancer patients.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein , Traitement néoadjuvant , Récepteur ErbB-3 , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/thérapie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/imagerie diagnostique , Traitement néoadjuvant/méthodes , Adulte d'âge moyen , Récepteur ErbB-3/métabolisme , Récepteur ErbB-3/génétique , Études prospectives , Adulte , Sujet âgé , Marqueurs biologiques tumoraux/métabolisme , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Récepteurs des oestrogènes/métabolisme , Régulation de l'expression des gènes tumoraux , Transduction du signal , Tomographie par émission de positons/méthodes
11.
J Immunol Res ; 2024: 3145695, 2024.
Article de Anglais | MEDLINE | ID: mdl-38983273

RÉSUMÉ

Background: This work focused on investigating the role of programmed death ligand 2 (PD-L2) in the progression of breast cancer by utilizing breast cancer specimens and cells. Materials and Methods: The serum levels of soluble PD-L2 (sPD-L2) in breast cancer patients and healthy individuals were analyzed by means of the enzyme-linked immunosorbent assay, and the PD-L2 levels within 416 resected breast cancer specimens were assessed through immunohistochemistry. Concurrently, in vitro cell experiments and in vivo animal experiments were carried out to analyze the relationship between PD-L2 and the invasion and migration of breast cancer. Results: The concentration of sPD-L2 in breast cancer patients significantly increased compared to that in the control groups. Additionally, breast cancer patients with high concentrations of sPD-L2 had higher Ki67 values (≥30%) and tumor grades. PD-L2 was expressed in 79.09% of the cancer samples, which exhibited a positive correlation with the progesterone receptor (PR) and the human epidermal growth factor receptor 2 (HER2). Furthermore, we discovered that knockdown of PD-L2 inhibited the migratory and invasive abilities of both MCF-7 and MDA-MB231 cells. Conclusion: Our findings demonstrated that knockdown of PD-L2 suppressed tumor growth, providing novel insights into important biological functions.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Évolution de la maladie , Ligand-2 de la protéine-1 de mort cellulaire programmée , Humains , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Animaux , Ligand-2 de la protéine-1 de mort cellulaire programmée/métabolisme , Ligand-2 de la protéine-1 de mort cellulaire programmée/génétique , Souris , Lignée cellulaire tumorale , Adulte d'âge moyen , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Adulte , Prolifération cellulaire , Cellules MCF-7 , Régulation de l'expression des gènes tumoraux , Invasion tumorale , Sujet âgé , Immunohistochimie , Grading des tumeurs , Marqueurs biologiques tumoraux/métabolisme , Modèles animaux de maladie humaine , Récepteurs à la progestérone/métabolisme , Techniques de knock-down de gènes
12.
Nat Commun ; 15(1): 5826, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992028

RÉSUMÉ

Patritumab deruxtecan (HER3-DXd) exhibits promising efficacy in breast cancer, with its activity not directly correlated to baseline ERBB3/HER3 levels. This research investigates the genetic factors affecting HER3-DXd's response in women with early-stage hormone receptor-positive and HER2-negative (HR+/HER2-) breast cancer. In the SOLTI-1805 TOT-HER3 trial, a single HER3-DXd dose was administered to 98 patients across two parts: 78 patients received 6.4 mg/kg (Part A), and 44 received a lower 5.6 mg/kg dose (Part B). The CelTIL score, measuring tumor cellularity and infiltrating lymphocytes from baseline to day 21, was used to assess drug activity. Part A demonstrated increased CelTIL score after one dose of HER3-DXd. Here we report CelTIL score and safety for Part B. In addition, the exploratory analyses of part A involve a comprehensive study of gene expression, somatic mutations, copy-number segments, and DNA-based subtypes, while Part B focuses on validating gene expression. RNA analyses show significant correlations between CelTIL responses, high proliferation genes (e.g., CCNE1, MKI67), and low expression of luminal genes (e.g., NAT1, SLC39A6). DNA findings indicate that CelTIL response is significantly associated with TP53 mutations, proliferation, non-luminal signatures, and a distinct DNA-based subtype (DNADX cluster-3). Critically, low HER2DX ERBB2 mRNA, correlates with increased HER3-DXd activity, which is validated through in vivo patient-derived xenograft  models. This study proposes chemosensitivity determinants, DNA-based subtype classification, and low ERBB2 expression as potential markers for HER3-DXd activity in HER2-negative breast cancer.


Sujet(s)
Anticorps monoclonaux humanisés , Tumeurs du sein , Récepteur ErbB-2 , Récepteur ErbB-3 , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Récepteur ErbB-3/métabolisme , Récepteur ErbB-3/génétique , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps neutralisants à large spectre/usage thérapeutique , Adulte d'âge moyen , Anticorps monoclonaux/usage thérapeutique , Adulte , Sujet âgé , Animaux , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Mutation , Souris , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Résultat thérapeutique , Trastuzumab , Camptothécine/analogues et dérivés , Immunoconjugués
13.
Cancer Med ; 13(14): e70035, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39031010

RÉSUMÉ

INTRODUCTION: The prognostic capability of targeted sequencing of primary tumors in patients with estrogen receptor-positive, human epidermal growth factor receptor-2-negative early-stage invasive breast cancer (EBC) in a real-world setting is uncertain. Therefore, we aimed to determine the correlation between a 22-gene mutational profile and long-term survival outcomes in patients with ER+/ERBB2- EBC. PATIENTS AND METHODS: A total of 73 women diagnosed with ER+/ERBB2- EBC between January 10, 2004, and June 2, 2008, were followed up until December 31, 2022. Univariate and multivariate Cox models were constructed to plot the relapse-free survival (RFS) and overall survival (OS). The log-rank test derived p-value was obtained. For external validation, we performed a survival analysis of 1163 comparable patients retrieved from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset. RESULTS: At follow-up, 16 (21.9%) patients had relapsed, while 21 (nearly 29%) harbored mutant genes. Thirty-three missense mutations were detected in 14 genes. The median ages were 51 and 46 years in patients with and without mutations, respectively. Patients with any mutation had a 1.85-fold higher risk of relapse (hazard ratio [HR]: 1.85, 95% confidence interval [CI]: 0.60-5.69) compared to those without any mutation. Patients who harbored any of the six genes (MAP2K4, FGFR3, APC, KIT, RB1, and PTEN) had a nearly 6-fold increase in the risk of relapse (HR: 5.82, 95% CI: 1.31-18.56; p = 0.0069). Multivariate Cox models revealed that the adjusted HR for RFS and OS were 6.67 (95% CI: 1.32-27.57) and 8.31 (p = 0.0443), respectively. METABRIC analysis also demonstrated a trend to significantly worse RFS (p = 0.0576) in the subcohort grouped by having a mutation in any of the six genes. CONCLUSIONS: Our single-institution tissue bank study of Taiwanese women with ER+/ERBB2- EBC suggests that a novel combination of six gene mutations might have prognostic capability for survival outcomes.


Sujet(s)
Tumeurs du sein , Mutation , Récepteur ErbB-2 , Récepteurs des oestrogènes , Humains , Femelle , Tumeurs du sein/génétique , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Adulte d'âge moyen , Récepteurs des oestrogènes/métabolisme , Pronostic , Adulte , Stadification tumorale , Marqueurs biologiques tumoraux/génétique , Sujet âgé , Invasion tumorale
14.
Sci Rep ; 14(1): 16899, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39043756

RÉSUMÉ

Mounting evidence showed that HER2-Low breast cancer patients could benefit from the novel anti-HER2 antibody-drug conjugates (ADCs) treatment, which pointed the way towards better therapy for HER2-Low patients. The purpose of this study was to describe the clinicopathological features, along with chemotherapeutic effects and survival outcomes of HER2-Low and HER2-Zero in TNBC who received neoadjuvant chemotherapy (NACT). We retrospectively evaluated 638 triple-negative breast cancer patients who were treated with neoadjuvant chemotherapy between August 2014 and August 2022. Pathologic complete response (pCR) and survival outcomes were analyzed in HER2-Low cohort, HER2-Zero cohort and the overall patients, respectively. In the entire cohort, 342 (53.6%) patients were HER2-Low and 296 (46.4%) patients were HER2-Zero. No significant difference was found between HER2-Low and HER2-Zero patients based on all the clinical-pathological characteristics. 143 cases (22.4%) achieved pCR after NACT in the overall TNBC patients. The pCR rate of the HER2-Low patients and the HER2-Zero patients was 21.3% and 23.6%, respectively, exhibiting no statistical difference (p = 0.487). The survival of pCR group after NACT significantly improved compared to non-pCR group either in HER2-Low patients or in HER2-Zero patients. Although we found that patients with HER2-Low had longer DFS than patients with HER2-Zero, there was no considerable difference (p = 0.068). However, HER2-Low patients had a dramatically longer OS than HER2-Zero patients (p = 0.012). The data from present study confirmed the clinical importance of HER2-Low expression in TNBC. Further effort is needed to determine whether HER2-Low could be a more favorable prognostic marker for individual treatment.


Sujet(s)
Traitement néoadjuvant , Récepteur ErbB-2 , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/mortalité , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Femelle , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Adulte d'âge moyen , Pronostic , Adulte , Études rétrospectives , Sujet âgé , Résultat thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique
15.
Nat Commun ; 15(1): 6130, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39033128

RÉSUMÉ

Cancer genomes are composed of many complex structural alterations on chromosomes and extrachromosomal DNA (ecDNA), making it difficult to identify non-coding enhancer regions that are hijacked to activate oncogene expression. Here, we describe a 3D genomics-based analysis called HAPI (Highly Active Promoter Interactions) to characterize enhancer hijacking. HAPI analysis of HiChIP data from 34 cancer cell lines identified enhancer hijacking events that activate both known and potentially novel oncogenes such as MYC, CCND1, ETV1, CRKL, and ID4. Furthermore, we found enhancer hijacking among multiple oncogenes from different chromosomes, often including MYC, on the same complex amplicons such as ecDNA. We characterized a MYC-ERBB2 chimeric ecDNA, in which ERBB2 heavily hijacks MYC's enhancers. Notably, CRISPRi of the MYC promoter led to increased interaction of ERBB2 with MYC enhancers and elevated ERBB2 expression. Our HAPI analysis tool provides a robust strategy to detect enhancer hijacking and reveals novel insights into oncogene activation.


Sujet(s)
Éléments activateurs (génétique) , Régulation de l'expression des gènes tumoraux , Génomique , Oncogènes , Régions promotrices (génétique) , Protéines proto-oncogènes c-myc , Récepteur ErbB-2 , Humains , Éléments activateurs (génétique)/génétique , Lignée cellulaire tumorale , Régions promotrices (génétique)/génétique , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Génomique/méthodes , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie
16.
Medicine (Baltimore) ; 103(24): e38508, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38875362

RÉSUMÉ

Breast cancer is currently the most commonly occurring cancer globally. Among breast cancer cases, the human epidermal growth factor receptor 2 (HER2)-positive breast cancer accounts for 15% to 20% and is a crucial focus in the treatment of breast cancer. Common HER2-targeted drugs approved for treating early and/or advanced breast cancer include trastuzumab and pertuzumab, which effectively improve patient prognosis. However, despite treatment, most patients with terminal HER2-positive breast cancer ultimately suffer death from the disease due to primary or acquired drug resistance. The prevalence of aberrantly activated the protein kinase B (AKT) signaling in HER2-positive breast cancer was already observed in previous studies. It is well known that p-AKT expression is linked to an unfavorable prognosis, and the phosphatidylinositol-3-kinase (PI3K)/AKT pathway, as the most common mutated pathway in breast cancer, plays a major role in the mechanism of drug resistance. Therefore, in the current review, we summarize the molecular alterations present in HER2-positive breast cancer, elucidate the relationships between HER2 overexpression and alterations in the PI3K/AKT signaling pathway and the pathways of the alterations in breast cancer, and summarize the resistant mechanism of drugs targeting the HER2-AKT pathway, which will provide an adjunctive therapeutic rationale for subsequent resistance to directed therapy in the future.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Protéines proto-oncogènes c-akt , Récepteur ErbB-2 , Transduction du signal , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Femelle , Protéines proto-oncogènes c-akt/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Phosphatidylinositol 3-kinases/métabolisme , Antinéoplasiques/usage thérapeutique , Phosphatidylinositol 3-kinase/métabolisme
18.
Biochem Biophys Res Commun ; 725: 150255, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-38897043

RÉSUMÉ

Human epidermal growth factor receptor 2 (HER2) aberrations are observed in various cancers. In non-small cell lung cancer, genetic alterations activating HER2, mostly exon 20 insertion mutations, occur in approximately 2-4% of cases. Trastuzumab deruxtecan (T-DXd), a HER2-targeted antibody-drug conjugate has been approved as the first HER2-targeted drug for HER2-mutant lung cancer. However, some cases are not responsive to T-DXd and the primary resistant mechanism remains unclear. In this study, we assessed sensitivity to T-DXd in JFCR-007, a patient-derived HER2-mutant lung cancer cell line. Although JFCR-007 was sensitive to HER2 tyrosine kinase inhibitors, it showed resistance to T-DXd in attachment or spheroid conditions. Accordingly, we established a three-dimensional (3D) layered co-culture model of JFCR-007, where it exhibited a lumen-like structure and became sensitive to T-DXd. In addition, an in-house inhibitor library screening revealed that G007-LK, a tankyrase inhibitor, was effective when combined with T-DXd. G007-LK increased the cytotoxicity of topoisomerase-I inhibitor, DXd, a payload of T-DXd and SN-38. This combined effect was also observed in H2170, an HER2-amplified lung cancer cell line. These results suggest that the proposed 3D co-culture system may help in evaluating the efficacy of T-DXd and may recapitulate the tumor microenvironment.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Techniques de coculture , Immunoconjugués , Tumeurs du poumon , Récepteur ErbB-2 , Trastuzumab , Humains , Trastuzumab/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Lignée cellulaire tumorale , Immunoconjugués/pharmacologie , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/antagonistes et inhibiteurs , Récepteur ErbB-2/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Éthers couronnes/pharmacologie , Antinéoplasiques immunologiques/pharmacologie , Camptothécine/analogues et dérivés
19.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891886

RÉSUMÉ

We present the case of a 70-year-old never-smoking female patient with epidermal growth factor receptor (EGFR) p.L858R-mutated metastatic non-small cell lung cancer (NSCLC). After three months of first-line treatment with erlotinib, progression occurred and platinum/pemetrexed was initiated, followed by a response for more than two years. After the progression, the molecular testing of a vertebral metastasis revealed a ROS proto-oncogene 1 (ROS1) translocation and a human epidermal growth factor receptor 2 (HER2) p.S310F mutation, in addition to the known EGFR p.L858R mutation. Crizotinib then led to a durable response of 17 months. The molecular retesting of the tumour cells obtained from the recurrent pleural effusion revealed the absence of the ROS1 translocation, whereas the EGFR and HER2 mutations were still present. Afatinib was added to the crizotinib, and the combination treatment resulted in another durable response of more than two years. The patient died more than 7 years after the initial diagnosis of metastatic NSCLC. This case demonstrates that the repeated molecular testing of metastatic NSCLC may identify new druggable genomic alterations that can impact the patient management and improve the patient outcome.


Sujet(s)
Adénocarcinome pulmonaire , Afatinib , Crizotinib , Récepteurs ErbB , Tumeurs du poumon , Protein-tyrosine kinases , Sujet âgé , Femelle , Humains , Adénocarcinome pulmonaire/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Afatinib/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Crizotinib/usage thérapeutique , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Mutation , Protein-tyrosine kinases/génétique , Protein-tyrosine kinases/métabolisme , Proto-oncogène Mas/génétique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme
20.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892097

RÉSUMÉ

Breast cancer remains a leading cause of cancer mortality in women globally. Despite advancements in systemic therapy, the risk of distant recurrence persists even after such treatment and may be linked to disseminated tumor cells (DTCs). Variability in molecular characteristics between primary tumors (PTs) and distant metastases underscores the need to comprehensively understand metastatic pathways. This retrospective study investigated discrepancies between HER2 expression in PTs and DTCs and their implications for survival outcomes in 201 early breast cancer (EBC) patients. We found a significant association between HER2 expression in PTs and DTCs when classifying tumors as HER2-high/low/negative. Patients whose HER2 status was discordant between PTs and DTCs exhibited worse distant disease-free survival than those with concordant status. Multivariate analysis confirmed the HER2 status of DTCs as an independent prognostic factor for distant DFS. These findings emphasize the importance of assessing HER2 expression in DTCs and its potential implications for tailored therapy strategies in EBC. Furthermore, prospective trials are needed to validate these findings and explore targeted therapies based on the molecular characteristics of DTCs.


Sujet(s)
Tumeurs du sein , Récepteur ErbB-2 , Humains , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Femelle , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/mortalité , Adulte d'âge moyen , Études rétrospectives , Adulte , Sujet âgé , Pronostic , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Survie sans rechute , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Métastase tumorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE