Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 15.204
Filtrer
1.
Mol Cancer ; 23(1): 169, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39164705

RÉSUMÉ

In the KEYNOTE-811 study, anti-HER2 and immunotherapy treatments resulted in longer survival in HER2-positive gastric cancer patients with CPS ≥ 1, whereas CPS < 1 patients lacked notable benefits. We studied this in a real-world cohort of 106 HER2-positive, CPS < 1 patients and found no survival differences between those treated with anti-HER2 therapy alone or with added immunotherapy. Thus, we investigate the tumor microenvironment variations in 160 HER2-positive patients, CPS ≥ 1 cases exhibited elevated spatial effective scores of immune cells, including CD4, CD8 subtypes, and NK cells, compared to CPS < 1. Furthermore, through single-cell sequencing in eight HER2-positive individuals, gene expressions revealed regulation of T-cell co-stimulation in CPS ≥ 1 and IL-1 binding in CPS < 1 cases. Notably, we discovered a CPS < 1 subtype marked by CXCR4+M2 macrophages, associated with poor prognosis, whose proportion and expression were reduced when benefiting from anti-HER2 therapy. These findings suggest CPS ≥ 1 patients, due to their immune microenvironment composition, may respond better to anti-PD-1/PD-L1 therapy.


Sujet(s)
Antigène CD274 , Récepteur ErbB-2 , Tumeurs de l'estomac , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/mortalité , Antigène CD274/métabolisme , Antigène CD274/génétique , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Pronostic , Marqueurs biologiques tumoraux , Régulation de l'expression des gènes tumoraux , Femelle , Immunothérapie/méthodes
2.
World J Surg Oncol ; 22(1): 214, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39164763

RÉSUMÉ

BACKGROUND: TCbHP (taxane + carboplatin + trastuzumab + pertuzumab) is the preferred neoadjuvant therapy regimen for human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, no consensus exists regarding whether specific populations may be exempt from carboplatin, allowing for de-escalation to the THP (taxane + trastuzumab + pertuzumab) regimen. Additionally, the optimal number of cycles for neoadjuvant THP remains unclear. We compared the efficacy and safety of neoadjuvant TCbHP and THP regimens, providing clinicians with a nuanced perspective to guide their treatment regimen selection. METHODS: This multicenter real-world study included patients with HER2-positive breast cancer undergoing neoadjuvant TCbHP or THP between March 2019 and February 2023. Efficacy was assessed through the pathological complete response (pCR) rate, while safety was evaluated through monitoring adverse events. RESULTS: Among 220 patients, 103 received 6 cycles of TCbHP (TCbHP×6), 83 received 6 cycles of THP (THP×6), and 34 received 4 cycles of THP (THP×4). The TCbHP×6 cohort exhibited a 66% pCR rate compared with 53% in the THP×6 cohort (P = 0.072). Subgroup analysis revealed that in patients aged ≤ 50 years, those with hormone receptor (HR)-negative status, and those with clinical stage T2, the pCR rate of the TCbHP×6 regimen was significantly higher than the THP×6 regimen (P < 0.05). The TCbHP×6 cohort reported higher frequencies of any-grade adverse events (99% versus 86.7%) and grade 3-4 events (49.5% versus 12%) than the THP×6 cohort. Propensity score matching identified 27 patient pairs between the THP×6 and THP×4 cohorts, indicating a significantly higher pCR rate for the THP×6 regimen than the THP×4 regimen (63% versus 29.6%, P = 0.029). CONCLUSIONS: The TCbHP×6 regimen is favored for individuals aged ≤ 50 years and those aged > 50, ≤60 years with HR-negative status or clinical stage T2-4. For patients in compromised general condition or lacking the specified indications, the THP×6 regimen emerges as a lower-toxicity alternative with satisfactory efficacy. To ensure treatment efficacy, a minimum of 6 cycles of neoadjuvant THP is required.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Carboplatine , Traitement néoadjuvant , Récepteur ErbB-2 , Taxoïdes , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Traitement néoadjuvant/méthodes , Carboplatine/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Récepteur ErbB-2/métabolisme , Adulte d'âge moyen , Chine/épidémiologie , Adulte , Taxoïdes/administration et posologie , Études de suivi , Trastuzumab/administration et posologie , Trastuzumab/usage thérapeutique , Pronostic , Études rétrospectives , Sujet âgé , Composés pontés/administration et posologie , Composés pontés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie
3.
J Exp Clin Cancer Res ; 43(1): 236, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39164784

RÉSUMÉ

BACKGROUND: Anti-HER2 therapies, including the HER2 antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd), have led to improved survival outcomes in patients with HER2-overexpressing (HER2+) metastatic breast cancer. However, intrinsic or acquired resistance to anti-HER2-based therapies remains a clinical challenge in these patients, as there is no standard of care following disease progression. The purpose of this study was to elucidate the mechanisms of resistance to T-DM1 and T-DXd in HER2+ BC patients and preclinical models and identify targets whose inhibition enhances the antitumor activity of T-DXd in HER2-directed ADC-resistant HER2+ breast cancer in vitro and in vivo. METHODS: Targeted DNA and whole transcriptome sequencing were performed in breast cancer patient tissue samples to investigate genetic aberrations that arose after anti-HER2 therapy. We generated T-DM1 and T-DXd-resistant HER2+ breast cancer cell lines. To elucidate their resistance mechanisms and to identify potential synergistic kinase targets for enhancing the efficacy of T-DXd, we used fluorescence in situ hybridization, droplet digital PCR, Western blotting, whole-genome sequencing, cDNA microarray, and synthetic lethal kinome RNA interference screening. In addition, cell viability, colony formation, and xenograft assays were used to determine the synergistic antitumor effect of T-DXd combinations. RESULTS: We found reduced HER2 expression in patients and amplified DNA repair-related genes in patients after anti-HER2 therapy. Reduced ERBB2 gene amplification in HER2-directed ADC-resistant HER2+ breast cancer cell lines was through DNA damage and epigenetic mechanisms. In HER2-directed ADC-resistant HER2+ breast cancer cell lines, our non-biased RNA interference screening identified the DNA repair pathway as a potential target within the canonical pathways to enhance the efficacy of T-DXd. We validated that the combination of T-DXd with ataxia telangiectasia and Rad3-related inhibitor, elimusertib, led to significant breast cancer cell death in vitro (P < 0.01) and in vivo (P < 0.01) compared to single agents. CONCLUSIONS: The DNA repair pathways contribute to HER2-directed ADC resistance. Our data justify exploring the combination treatment of T-DXd with DNA repair-targeting drugs to treat HER2-directed ADC-resistant HER2+ breast cancer in clinical trials.


Sujet(s)
Tumeurs du sein , Réparation de l'ADN , Résistance aux médicaments antinéoplasiques , Immunoconjugués , Récepteur ErbB-2 , Trastuzumab , Humains , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Trastuzumab/pharmacologie , Trastuzumab/usage thérapeutique , Animaux , Immunoconjugués/pharmacologie , Immunoconjugués/usage thérapeutique , Souris , Récepteur ErbB-2/métabolisme , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Camptothécine/analogues et dérivés , Camptothécine/pharmacologie , Camptothécine/usage thérapeutique , Synergie des médicaments
4.
Breast Cancer Res ; 26(1): 125, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39192322

RÉSUMÉ

In many countries, hormone receptor status assessment of ductal carcinoma in situ (DCIS) is routinely performed, as hormone receptor-positive DCIS patients are eligible for adjuvant anti-hormonal treatment, aiming to reduce the ipsilateral and contralateral breast cancer risk. Although HER2 gene amplification and its associated HER2 protein overexpression constitute a major prognostic and predictive marker in invasive breast carcinoma, its use in the diagnosis and treatment of DCIS is less straightforward. HER2 immunohistochemistry is not routinely performed yet, as the role of HER2-positivity in DCIS biology is unclear. Nonetheless, recent data challenge this practice. Here, we discuss the value of routine HER2 assessment for DCIS. HER2-positivity correlates strongly with DCIS grade: around four in five HER2-positive DCIS show high grade atypia. As morphological DCIS grading is prone to interobserver variability, HER2 immunohistochemistry could render grading more robust. Several studies showed an association between HER2-positive DCIS and ipsilateral recurrence risk, albeit currently unclear whether this is for overall, in situ or invasive recurrence. HER2-positive DCIS tends to be larger, with a higher risk of involved surgical margins. HER2-positive DCIS patients benefit more from adjuvant radiotherapy: it substantially decreases the local recurrence risk after lumpectomy, without impact on overall survival. HER2-positivity in pure biopsy-diagnosed DCIS is associated with increased upstaging to invasive carcinoma after surgery. HER2 immunohistochemistry on preoperative biopsies might therefore provide useful information to surgeons, favoring wider excisions. The time seems right to consider DCIS subtype-dependent treatment, comprising appropriate local treatment for HER2-positive DCIS patients and de-escalation for hormone receptor-positive, HER2-negative DCIS patients.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein , Carcinome intracanalaire non infiltrant , 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/thérapie , Tumeurs du sein/mortalité , Tumeurs du sein/diagnostic , Carcinome intracanalaire non infiltrant/anatomopathologie , Carcinome intracanalaire non infiltrant/thérapie , Carcinome intracanalaire non infiltrant/métabolisme , Carcinome intracanalaire non infiltrant/génétique , Marqueurs biologiques tumoraux/métabolisme , Pronostic , Immunohistochimie , Récidive tumorale locale/métabolisme , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/génétique , Grading des tumeurs , Pertinence clinique
5.
J Biosci ; 492024.
Article de Anglais | MEDLINE | ID: mdl-39193850

RÉSUMÉ

Circadian clocks, biochemical oscillators that are regulated by environmental time cues including the day/night cycle, have a central function in the majority of biological processes. The disruption of the circadian clock can alter breast biology negatively and may promote the development of breast tumors. The expression status of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) were used to classify breast cancer into different molecular subtypes such as triple-negative breast cancer (TNBC). Receptor status-dependent expression of circadian clock genes have been previously studied in breast cancer using relatively small sample sizes in a particular population. Here, using TCGA-BRCA data (n=1119), we found that the expressions of CRY1, PER1, PER2, PER3, BMAL1, CLOCK, RORA, RORB, RORC, NR1D1, NR1D2, and FBXL3 were higher in ER+ breast cancer cells compared with those of ER- status. Similarly, we showed that transcript levels of CRY2, PER1, PER2, PER3, BMAL1, RORA, RORB, RORC, NR1D1, NR1D2, and FBXL3 were higher in PR+ breast cancer cells than in PR- breast cancer cells. We report that the expressions of CRY2, PER1, BMAL1, and RORA were lower, and the expression of NR1D1 was higher, in HER2+ breast cancer cells compared with HER2- breast cancer cells. Moreover, we studied these receptor status-dependent changes in the expressions of circadian clock genes also based on the race and age of breast cancer patients. Lastly, we found that the expressions of CRY2, PER1, PER2, PER3, and CLOCK were higher in non-TNBC than in TNBC, which has the worst prognosis among subtypes. We note that our findings are not always parallel to the observations reported in previous studies with smaller sample sizes performed in different populations and organisms. Our study suggests that receptor status in breast cancer (thus, subtype of breast cancer) might be more important than previously shown in terms of its influence on the expression of circadian clock genes and on the disruption of the circadian clock, and that ER or PR might be important regulators of breast cancer chronobiology that should be taken into account in personalized chronotherapies.


Sujet(s)
Tumeurs du sein , Horloges circadiennes , Régulation de l'expression des gènes tumoraux , Récepteurs des oestrogènes , Récepteurs à la progestérone , Humains , Femelle , Récepteurs à la progestérone/métabolisme , Récepteurs à la progestérone/génétique , Horloges circadiennes/génétique , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Lignée cellulaire tumorale , Cryptochromes/génétique , Cryptochromes/métabolisme
6.
Curr Oncol ; 31(8): 4728-4745, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39195336

RÉSUMÉ

Despite consensus guidelines, most patients with early-stage triple-negative (TN) and HER2-positive (HER2+) breast cancer do not see a medical oncologist prior to surgery and do not receive neoadjuvant chemotherapy (NAC). To understand barriers to care, we aimed to characterize the relationship between geography (region of residence and cancer centre proximity) and receipt of a pre-treatment medical oncology consultation and NAC for patients with TN and HER2+ breast cancer. Using linked administrative datasets in Ontario, Canada, we performed a retrospective population-based analysis of women diagnosed with stage I-III TN or HER2+ breast cancer from 2012 to 2020. The outcomes were a pre-treatment medical oncology consultation and the initiation of NAC. We created choropleth maps to assess the distribution of the outcomes and cancer centres across census divisions. To assess the relationship between distance to the nearest cancer centre and outcomes, we performed multivariable regression analyses adjusted for relevant factors, including tumour extent and nodal status. Of 14,647 patients, 29.9% received a pre-treatment medical oncology consultation and 77.7% received NAC. Mapping demonstrated high interregional variability, ranging across census divisions from 12.5% to 64.3% for medical oncology consultation and from 8.8% to 64.3% for NAC. In the full cohort, compared to a distance of ≤5 km from the nearest cancer centre, only 10-25 km was significantly associated with lower odds of NAC (OR 0.83, 95% CI 0.70-0.99). Greater distances were not associated with pre-treatment medical oncology consultation. The interregional variability in medical oncology consultation and NAC for patients with TN and HER2+ breast cancer suggests that regional and/or provider practice patterns underlie discrepancies in the referral for and receipt of NAC. These findings can inform interventions to improve equitable access to NAC for eligible patients.


Sujet(s)
Traitement néoadjuvant , Orientation vers un spécialiste , Tumeurs du sein triple-négatives , Humains , Femelle , Traitement néoadjuvant/méthodes , Traitement néoadjuvant/statistiques et données numériques , Adulte d'âge moyen , Tumeurs du sein triple-négatives/traitement médicamenteux , Études rétrospectives , Orientation vers un spécialiste/statistiques et données numériques , Adulte , Sujet âgé , Ontario , Récepteur ErbB-2/métabolisme , Oncologie médicale/statistiques et données numériques , Oncologie médicale/méthodes , Accessibilité des services de santé/statistiques et données numériques , Tumeurs du sein/traitement médicamenteux , Établissements de cancérologie/statistiques et données numériques
7.
Physiol Rep ; 12(16): e70008, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39187401

RÉSUMÉ

The irreversible pan-HER tyrosine kinase inhibitor neratinib is approved for patients with HER2-positive, early-stage and metastatic breast cancer (BC). Neratinib-associated diarrhea is the most common reason for early discontinuation. Preclinical studies identified mechanisms of neratinib-induced diarrhea and rationale for prophylactic and preventive measures. We studied effects of neratinib on rat intestines and conducted a phase 2 study of colon pathogenesis in patients with HER2-positive BC treated with neratinib (NCT04366713). Colon samples from female albino Wistar rats receiving neratinib or vehicle were examined for histopathological changes. Patients with HER2-positive BC received neratinib 240 mg once daily for up to 1 year. Colonoscopy biopsies were collected at baseline and at Day 28 to identify changes consistent with rat pathologies. Rat colons were markedly altered in appearance, with similar short circuit currents (Isc) and responses to carbachol and forskolin. Mucosal barrier loss and/or significant increase in secretory propensity in neratinib- versus control-treated animals were not seen. Two of four endpoint-evaluable patients presented with mild pathological changes, largely comparable with the rat model. Preclinical evidence supports an inflammatory component of neratinib-induced diarrhea without mucosal barrier function loss. Colonoscopy findings in patients with BC indicate mild or no pathological changes in the colon due to neratinib treatment.


Sujet(s)
Tumeurs du sein , Coloscopie , Diarrhée , Quinoléines , Rat Wistar , Récepteur ErbB-2 , Animaux , Femelle , Diarrhée/induit chimiquement , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Rats , Projets pilotes , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Humains , Adulte d'âge moyen , Côlon/anatomopathologie , Côlon/effets des médicaments et des substances chimiques , Côlon/métabolisme , Sujet âgé , Antinéoplasiques/effets indésirables , Antinéoplasiques/toxicité
8.
Sci Rep ; 14(1): 18118, 2024 08 05.
Article de Anglais | MEDLINE | ID: mdl-39103402

RÉSUMÉ

Breast cancer is among the highest morbidity and mortality rates in women around the world. In the present investigation we aimed to synthesis novel nanosystem combining two naturally important anticancer agents with different mechanism of action namely Moringa oleifera and caffeine. Firstly, chemical analysis of Moringa oleifera extract and caffeine was done by gas chromatography-mass spectroscopy (GC-MS) in order to assess the main chemical compounds present and correlate between them and the possible anticancer effect. The novel nanosystem was characterized through dynamic light scattering techniques which revealed the stability and homogeneity of the prepared M. oleifera leaves extract/Caffeine loaded chitosan nanoparticles, while FTIR and transmission electron microscope (TEM) proved the shape and the successful incorporation of M. oleifera leaves extract/Caffeine onto the nanochitosan carrier. Our initial step was to assess the anticancer effect in vitro in cancer cell line MCF-7 which proved the significant enhanced effect of M. oleifera leaves extract/Caffeine nanosystem compared to M. oleifera leaves extract or caffeine loaded nanoparticles. Further studies were conducted in vivo namely tumor biomarkers, tumor volume, bioluminescence imaging, molecular and histopathological investigations. The present study proved the potent anticancer effect of the synthesized M. oleifera leaves extract/Caffeine loaded chitosan nanoparticles. Mo/Caf/CsNPs exhibited a large number of apoptotic cells within the tumor mass while the adipose tissue regeneration was higher compared to the positive control. The prepared nanoparticles downregulated the expression of Her2, BRCA1 and BRCA2 while mTOR expression was upregulated. The aforementioned data demonstrated the successful synergistic impact of Moringa and caffeine in decreasing the carcinoma grade.


Sujet(s)
Protéine BRCA1 , Protéine BRCA2 , Tumeurs du sein , Caféine , Chitosane , Nanoparticules , Extraits de plantes , Feuilles de plante , Récepteur ErbB-2 , Chitosane/composition chimique , Humains , Caféine/pharmacologie , Caféine/composition chimique , Nanoparticules/composition chimique , Feuilles de plante/composition chimique , Femelle , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Cellules MCF-7 , Protéine BRCA2/génétique , Protéine BRCA2/métabolisme , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Protéine BRCA1/génétique , Protéine BRCA1/métabolisme , Animaux , Moringa oleifera/composition chimique , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
10.
BMC Cancer ; 24(1): 952, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097705

RÉSUMÉ

BACKGROUND: Limited data exists on the efficacy of immune checkpoint inhibitor (ICI) combinations in non-small-cell lung cancer (NSCLC) with uncommon driver alterations in genes such as ERBB2, BRAF, RET, and MET. This study retrospectively assessed ICI-combination therapy outcomes in this molecular subset of NSCLC. METHODS: We retrospectively analyzed patients with advanced NSCLC confirmed with driver alterations in genes including ERBB2, BRAF, RET or MET, and received ICI combined with chemotherapy (ICI + chemo) and/or targeted therapy (ICI + chemo/TT) as first-line (1L) or second- or third-line (≥ 2L) treatment at Hunan Cancer Hospital between January 2018 and May 2024. RESULTS: Of the 181 patients included in the study, 131 patients received 1L-ICI + chemo (ERBB2, n = 64; BRAF, n = 34; RET, n = 23; and MET, n = 10), and 50 patients received ≥ 2L-ICI + chemo/TT (ERBB2, n = 16; BRAF, n = 7; RET, n = 14; MET, n = 13). The full cohort had an overall response rate (ORR) of 45.9% and disease control rate of 84.0%. Among patients who received 1L-ICI + chemo, ORR ranged between 51.6% and 60.0%, with the median progression-free survival (mPFS) and overall survival (mOS) of 8.2 and 21.0 months for those with ERBB2-altered tumors, 10.0 and 15.0 months for BRAF-altered tumors, 12.1 months and OS not reached for RET-altered tumors, and 6.2 and 28.0 months for MET-altered tumors, respectively. Additionally, ORR ranged between 14.3% and 30.8% for ≥ 2L-ICI + chemo/TT; mPFS and mOS were 5.4 and 16.2 months for patients with ERBB2-altered tumors, 2.7 and 5.0 months for BRAF-altered tumors, 6.2 and 14.3 months for RET-altered tumors, and 5.7 and 11.5 months for MET-altered tumors, respectively. CONCLUSION: ICI-based combination therapies, regardless of treatment line, were effective in treating patients with advanced NSCLC harboring driver alterations in ERBB2, BRAF, RET, or MET. This suggests their potential as alternative treatment options in this patient population.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Carcinome pulmonaire non à petites cellules , Inhibiteurs de points de contrôle immunitaires , Tumeurs du poumon , Récepteur ErbB-2 , 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 , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Adulte d'âge moyen , Sujet âgé , Études rétrospectives , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte , Récepteur ErbB-2/génétique , Récepteur ErbB-2/métabolisme , Sujet âgé de 80 ans ou plus , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes c-ret/génétique , Résultat thérapeutique , Mutation , Protéines proto-oncogènes c-met/génétique
12.
Rev Med Inst Mex Seguro Soc ; 62(1): 1-7, 2024 Jan 08.
Article de Espagnol | MEDLINE | ID: mdl-39110839

RÉSUMÉ

Background: Primary breast tumors with neuroendocrine (NE) differentiation are a heterogeneous tumor group with diversity of biological behavior, with poorly defined prevalence and prognosis. Objective: To evaluate the chromogranin, synaptophysin, CD56, INSM1 markers expression prevalence and the association between NE differentiation and tumor molecular type. Material and methods: Observational, cross-sectional study which included 110 breast tissue samples with primary invasive carcinoma. Immunohistochemistry was performed for chromogranin, synaptophysin, CD56 and INMS1 markers. NE differentiation was considered with 10-90% positive cells, and NE tumor with > 90% positive cells. Results: 26.3% showed neuroendocrine differentiation. Out of these, 48.2% were luminal-A type, 24.1% luminal-B, 11.5% HER2neu, 17.2% triple-negative; 1.8% were NE tumors. Tumors were marker positive, and out of these to chromogranin in 24.5%, synaptophysin in 28.2%, CD56 in 2.7%, INSM1 in 16.4%. Synaptophysin was expressed in 17.3% luminal-A type, 6.4% luminal-B, 0.9% HER2neu, 3.6% triple-negative. NE differentiation showed association with synaptophysin expression (r = 0.586, p = 0.0001). Conclusion: The NE differentiation prevalence was 26.3% in primary invasive breast cancers, with luminal-A molecular type predominance.


Introducción: los tumores primarios de mama con diferenciación neuroendócrina (NEBC por sus siglas en inglés) son un grupo heterogéneo de tumores con diversidad de comportamiento biológico, con prevalencia y pronóstico poco definido. Objetivo: evaluar la prevalencia de la expresión los marcadores cromogranina, sinaptofisina, CD56, INSM1 y la asociación entre la diferenciación neuroendócrina y el tipo molecular del tumor. Material y métodos: estudio observacional, transversal que incluyó 110 muestras de tejido mamario con carcinoma invasor primario. Se realizó inmunohistoquímica para los marcadores cromogranina, sinaptofisina, CD56 y INMS1. La presencia 10-90% de células positivas se consideró diferenciación neuroendócrina y tumor neuroendócrino con > 90% de células positivas. Resultados: el 26.3% mostró diferenciación neuroendócrina. De estos, 48.2% fueron tipo luminal-A, 24.1% luminal-B, 11.5% HER2neu y 17.2% triple-negativo; 1.8% resultaron tumores neuroendócrinos. Los tumores presentaron marcadores positivos y de estos, 24.5% fueron a cromogranina, 28.2% a sinaptofisina, 2.7% a CD56 y 16.4% a INSM1. La sinaptofisina se expresó en 17.3% del tipo luminal-A, 6.4% luminal-B, 0.9% HER2neu, 3.6% triple-negativo. La diferenciación neuroendócrina mostró asociación con la expresión de sinaptofisina (r = 0.586, p = 0.0001). Conclusión: la prevalencia de la diferenciación neuroendócrina fue del 26.3% en los cánceres invasores primarios de mama, con predominio en el tipo molecular luminal-A.


Sujet(s)
Marqueurs biologiques tumoraux , Synaptophysine , Humains , Femelle , Études transversales , Marqueurs biologiques tumoraux/métabolisme , Adulte d'âge moyen , Adulte , Synaptophysine/métabolisme , Sujet âgé , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Antigènes CD56/métabolisme , Immunohistochimie , Protéines de répression/métabolisme , Chromogranine/métabolisme , Récepteur ErbB-2/métabolisme , Sujet âgé de 80 ans ou plus
13.
J Transl Med ; 22(1): 745, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39113057

RÉSUMÉ

BACKGROUND: Human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC), which accounts for approximately one-fifth of all BCs, are highly invasive with a high rate of recurrence and a poor prognosis. Several studies have shown that growth factor receptor-bound protein 7 (GRB7) might be a potential therapeutic target for tumor diagnosis and prognosis. Nevertheless, the role of GRB7 in HER2+ BC and its underlying mechanisms have not been fully elucidated. The aim of this study was to investigate the biological function and regulatory mechanism of GRB7 in HER2+ BC. METHODS: Bioinformatics analysis was performed using the TCGA, GEO and CancerSEA databases to evaluate the clinical significance of GRB7. RT quantitative PCR, western blot and immunofluorescence were conducted to assess the expression of GRB7 in BC cell lines and tissues. MTT, EdU, colony formation, wound healing, transwell, and xenograft assays were adopted to explore the biological function of GRB7 in HER2+ BC. RNA sequencing was performed to analyze the signaling pathways associated with GRB7 in SK-BR-3 cells after the cells were transfected with GRB7 siRNA. Chromatin immunoprecipitation analysis (ChIP) and luciferase reporter assay were employed to elucidate the potential molecular regulatory mechanisms of GRB7 in HER2+ BC. RESULTS: GRB7 was markedly upregulated and associated with poor prognosis in BC, especially in HER2+ BC. Overexpression of GRB7 increased the proliferation, migration, invasion, and colony formation of HER2+ BC cells, while depletion of GRB7 had the opposite effects in HER2+ BC cells and inhibited xenograft growth. ChIP-PCR and luciferase reporter assay revealed that TCF12 directly bound to the promoter of the GRB7 gene to promote its transcription. GRB7 facilitated HER2+ BC epithelial-mesenchymal transition (EMT) progression by interacting with Notch1 to activate Wnt/ß-catenin pathways and other signaling (i.e., AKT, ERK). Moreover, forced GRB7 overexpression activated Wnt/ß-catenin to promote EMT progression, and partially rescued the inhibition of HER2+ BC proliferation, migration and invasion induced by TCF12 silencing. CONCLUSIONS: Our work elucidates the oncogenic role of GRB7 in HER2+ BC, which could serve as a prognostic indicator and promising therapeutic target.


Sujet(s)
Tumeurs du sein , Prolifération cellulaire , Évolution de la maladie , Protéine adaptatrice GRB7 , Régulation de l'expression des gènes tumoraux , Récepteur ErbB-2 , Récepteur Notch1 , Transduction du signal , Humains , Protéine adaptatrice GRB7/métabolisme , Protéine adaptatrice GRB7/génétique , Femelle , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Animaux , Récepteur Notch1/métabolisme , Récepteur Notch1/génétique , Souris nude , Mouvement cellulaire/génétique , Transition épithélio-mésenchymateuse/génétique , Souris , Invasion tumorale , Souris de lignée BALB C , Facteurs de transcription à motif basique hélice-boucle-hélice
14.
Cancer Immunol Immunother ; 73(10): 197, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39105849

RÉSUMÉ

BACKGROUND: Biomarkers for predicting response to the immunotherapy and chemotherapy combination in breast cancer patients are not established. In this study, we report exploratory genomic and transcriptomic analyses of pretreatment tumor tissues from patients enrolled in phase II clinical trial of a combination of eribulin and nivolumab for HER-2-negative metastatic breast cancer (MBC) (KORNELIA trial, NCT04061863). METHODS: We analyzed associations between tumor molecular profiles based on genomic (n = 76) and transcriptomic data (n = 58) and therapeutic efficacy. Patients who achieved progression-free survival (PFS) ≥ 6 months were defined as PFS6-responders and PFS6-nonresponders otherwise. FINDINGS: Analyses on tumor mutation burden (TMB) showed a tendency toward a favorable effect on efficacy, while several analyses related to homologous recombination deficiency (HRD) indicated a potentially negative impact on efficacy. Patients harboring TP53 mutations showed significantly poor PFS6 rate and PFS, which correlated with the enrichment of cell cycle-related signatures in PFS6-nonresponders. High antigen presentation gene set enrichment scores (≥ median) were significantly associated with longer PFS. Naïve B-cell and plasma cell proportions were considerably higher in long responders (≥ 18 months). INTERPRETATION: Genomic features including TMB, HRD, and TP53 mutations and transcriptomic features related to immune cell profiles and cell cycle may distinguish responders. Our findings provide insights for further exploring the combination regimen and its biomarkers in these tumors.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Furanes , Cétones , Nivolumab , Récepteur ErbB-2 , Transcriptome , Humains , Femelle , Cétones/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/mortalité , Furanes/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Nivolumab/usage thérapeutique , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Adulte d'âge moyen , Génomique/méthodes , Sujet âgé , Marqueurs biologiques tumoraux/génétique , Adulte , Mutation , Métastase tumorale , Analyse de profil d'expression de gènes ,
15.
BMC Oral Health ; 24(1): 898, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107736

RÉSUMÉ

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) plays an important role in the progression of multiple solid tumors and induces resistance to epidermal growth factor receptor (EGFR) target treatment. However, the expression status and the clinical significance of HER2 in oral squamous cell carcinoma (OSCC) is still controversial. Pyrotinib (PYR) is a promising novel EGFR/HER2 dual inhibitor, whose efficacy in OSCC has not been determined. METHODS: 57 locally advanced de novo OSCC patients were included in this study to investigate the relationship between the HER2 expression levels and the prognosis by the tissue microarray analysis (TMA). In vitro and in vivo experiments were performed to retrieve the efficacy of PYR in OSCC. The main downstream of HER2 was evaluated by western blotting in OSCC cell lines and xenograft tumors to explore the potential mechanism of PYR. RESULTS: This study revealed the primary tumor of OSCC had higher HER2 expression levels. Patients with HER2 overexpression had poor overall survival (P < 0.014) and poor disease free survival (P < 0.042). In vitro, PYR suppressed the proliferation, colony formation and migration of OSCC cells. It also promoted apoptosis of OSCC cells and induced cell cycle arrest. Furthermore, PYR was able to inhibit the occurrence and development of OSCC effectively in vivo. Western blotting revealed that PYR suppressed OSCC by inhibiting the phosphorylation of HER2, AKT and ERK. CONCLUSIONS: This study exhibited the anti-OSCC effects of PYR in vitro and in vivo, and demonstrated PYR inhibited OSCC cells by inducing apoptosis via the HER2/ AKT and ERK pathway. The result of this study also indicated locally advanced OSCC patients might benefit from HER2 assay and EGFR/HER2 dual inhibit treatment.


Sujet(s)
Carcinome épidermoïde , Récepteurs ErbB , Tumeurs de la bouche , Récepteur ErbB-2 , Humains , Tumeurs de la bouche/traitement médicamenteux , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Récepteur ErbB-2/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Animaux , Lignée cellulaire tumorale , Femelle , Souris , Mâle , Apoptose/effets des médicaments et des substances chimiques , Acrylamides/pharmacologie , Acrylamides/usage thérapeutique , Adulte d'âge moyen , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Aminoquinoléines
16.
BMC Cancer ; 24(1): 991, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39128984

RÉSUMÉ

BACKGROUND: Our previous study demonstrated that ß2-microglobulin (ß2M) promoted ER+/HER2- breast cancer survival via the SGK1/Bcl-2 signaling pathway. However, the role of ß2M has not been investigated in ER-/HER2+ breast cancer. Here, we aimed to determine the role of ß2M in ER-/HER2+ breast cancer. METHODS: The interaction between ß2M and HFE was confirmed by co-immunoprecipitation, mass spectrometry, yeast two-hybrid screening, and His pull-down. The knockdown and overexpression of ß2M or HFE were performed in MDA-MB-453 cells, and ERK signaling pathway was subsequently analyzed via western blotting. Apoptotic cells were detected using flow cytometer. ß2M, HFE, and p-ERK1/2 were examined in tumor and paired adjacent tissues via immunohistochemistry. RESULTS: HFE was found to be an interacting protein of ß2M in ER-/HER2+ breast cancer cells MDA-MB-453 by co-immunoprecipitation and mass spectrometry. A yeast two-hybrid system and His-pull down experiments verified that ß2M directly interacted with HFE. ß2M and HFE as a complex were mainly located in the cytoplasm, with some on the cytomembrane of MDA-MB-453 cells. In addition to breast cancer cells BT474, endogenous ß2M directly interacted with HFE in breast cancer cells MDA-MB-453, MDA-MB-231, and MCF-7. ß2M activated the ERK signaling pathway by interacting with HFE and induced apoptosis of MDA-MB-453 cells. The expression of HFE and p-ERK1/2 showed significantly high levels in HER2-overexpressing breast cancer tumor tissue compared with adjacent normal tissue, consistent with the results obtained from the cell experiments. CONCLUSIONS: ß2M induced apoptosis of tumor cells via activation of the ERK signal pathway by directly interacting with HFE in HER2-overexpressing breast cancer.


Sujet(s)
Apoptose , Tumeurs du sein , Protéine de l'hémochromatose , Système de signalisation des MAP kinases , Récepteur ErbB-2 , bêta-2-Microglobuline , Humains , bêta-2-Microglobuline/métabolisme , bêta-2-Microglobuline/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Femelle , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Lignée cellulaire tumorale , Protéine de l'hémochromatose/génétique , Protéine de l'hémochromatose/métabolisme , Liaison aux protéines , Régulation de l'expression des gènes tumoraux
17.
Drug Resist Updat ; 76: 101118, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39094301

RÉSUMÉ

AIMS: Resistance to targeted therapy is one of the critical obstacles in cancer management. Resistance to trastuzumab frequently develops in the treatment for HER2+ cancers. The role of protein tyrosine phosphatases (PTPs) in trastuzumab resistance is not well understood. In this study, we aim to identify pivotal PTPs affecting trastuzumab resistance and devise a novel counteracting strategy. METHODS: Four public datasets were used to screen PTP candidates in relation to trastuzumab responsiveness in HER2+ breast cancer. Tyrosine kinase (TK) arrays were used to identify kinases that linked to protein tyrosine phosphate receptor type O (PTPRO)-enhanced trastuzumab sensitivity. The efficacy of small activating RNA (saRNA) in trastuzumab-conjugated silica nanoparticles was tested for PTPRO upregulation and resistance mitigation in cell models, a transgenic mouse model, and human cancer cell line-derived xenograft models. RESULTS: PTPRO was identified as the key PTP which influences trastuzumab responsiveness and patient survival. PTPRO de-phosphorated several TKs, including the previously overlooked substrate ERBB3, thereby inhibiting multiple oncogenic pathways associated with drug resistance. Notably, PTPRO, previously deemed "undruggable," was effectively upregulated by saRNA-loaded nanoparticles. The upregulated PTPRO simultaneously inhibited ERBB3, ERBB2, and downstream SRC signaling pathways, thereby counteracting trastuzumab resistance. CONCLUSIONS: Antibody-conjugated saRNA represents an innovative approach for targeting "undruggable" PTPs.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Nanoparticules , Récepteur ErbB-2 , Trastuzumab , Tests d'activité antitumorale sur modèle de xénogreffe , Trastuzumab/pharmacologie , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Animaux , Souris , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Femelle , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Nanoparticules/composition chimique , Souris transgéniques , Antinéoplasiques immunologiques/pharmacologie , Receptor-Like Protein Tyrosine Phosphatases, Class 3/métabolisme , Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonistes et inhibiteurs , Receptor-Like Protein Tyrosine Phosphatases, Class 3/génétique , Transduction du signal/effets des médicaments et des substances chimiques
18.
Nat Commun ; 15(1): 6587, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097623

RÉSUMÉ

Metabolic reprogramming, a hallmark of tumorigenesis, involves alterations in glucose and fatty acid metabolism. Here, we investigate the role of Carnitine palmitoyl transferase 1a (Cpt1a), a key enzyme in long-chain fatty acid (LCFA) oxidation, in ErbB2-driven breast cancers. In ErbB2+ breast cancer models, ablation of Cpt1a delays tumor onset, growth, and metastasis. However, Cpt1a-deficient cells exhibit increased glucose dependency that enables survival and eventual tumor progression. Consequently, these cells exhibit heightened oxidative stress and upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) activity. Inhibiting Nrf2 or silencing its expression reduces proliferation and glucose consumption in Cpt1a-deficient cells. Combining the ketogenic diet, composed of LCFAs, or an anti-ErbB2 monoclonal antibody (mAb) with Cpt1a deficiency significantly perturbs tumor growth, enhances apoptosis, and reduces lung metastasis. Using an immunocompetent model, we show that Cpt1a inhibition promotes an antitumor immune microenvironment, thereby enhancing the efficacy of anti-ErbB2 mAbs. Our findings underscore the importance of targeting fatty acid oxidation alongside HER2-targeted therapies to combat resistance in HER2+ breast cancer patients.


Sujet(s)
Tumeurs du sein , Carnitine O-palmitoyltransferase , Acides gras , Facteur-2 apparenté à NF-E2 , Oxydoréduction , Récepteur ErbB-2 , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Récepteur ErbB-2/antagonistes et inhibiteurs , Acides gras/métabolisme , Carnitine O-palmitoyltransferase/métabolisme , Carnitine O-palmitoyltransferase/génétique , Animaux , Femelle , Humains , Souris , Lignée cellulaire tumorale , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Stress oxydatif , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Régime cétogène , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Glucose/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/secondaire , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie
19.
Commun Biol ; 7(1): 983, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138287

RÉSUMÉ

The mechanism of action of bispecific antibodies (bsAbs) directing T-cell immunity to solid tumors is incompletely understood. Here, we screened a series of CD3xHER2 bsAbs using extracellular matrix (ECM) embedded breast cancer tumoroid arrays exposed to healthy donor-derived T-cells. An initial phase of random T-cell movement throughout the ECM (day 1-2), was followed by a bsAb-dependent phase of active T-cell recruitment to tumoroids (day 2-4), and tumoroid killing (day 4-6). Low affinity HER2 or CD3 arms were compensated for by increasing bsAb concentrations. Instead, a bsAb binding a membrane proximal HER2 epitope supported tumor killing whereas a bsAb binding a membrane distal epitope did not, despite similar affinities and intra-tumoroid localization of the bsAbs, and efficacy in 2D co-cultures. Initial T-cell-tumor contact through effective bsAbs triggered a wave of subsequent T-cell recruitment. This critical surge of T-cell recruitment was explained by paracrine signaling and preceded a full-scale T-cell tumor attack.


Sujet(s)
Anticorps bispécifiques , Antigènes CD3 , Communication paracrine , Lymphocytes T , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/immunologie , Humains , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Lymphocytes T/immunologie , Femelle , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Récepteur ErbB-2/immunologie , Récepteur ErbB-2/métabolisme , Lignée cellulaire tumorale
20.
Cancer Med ; 13(15): e7408, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39136200

RÉSUMÉ

BACKGROUND: The MONALEESA­7 and ­2 phase 3 randomized trials demonstrated a statistically significant progression­free survival (PFS) and overall survival (OS) benefit with initial ribociclib + endocrine therapy (ET) versus placebo + ET in pre­ and postmenopausal patients with hormone receptor­positive (HR+)/human epidermal growth factor receptor 2­negative (HER2−) advanced breast cancer (ABC), respectively. Similar trends were observed in Asian subgroup analyses. This phase 2 bridging study of initial ET + ribociclib enrolled pre­ and postmenopausal patients with HR+/HER2­ ABC from China and was conducted to demonstrate consistency of PFS results in a Chinese population relative to the global MONALEESA­7 and ­2 studies. METHODS: Patients were randomized (1:1) to ET (nonsteroidal aromatase inhibitor + goserelin for premenopausal patients; letrozole for postmenopausal patients) + either ribociclib or placebo. The primary endpoint was investigator­assessed PFS. RESULTS: As of April 25, 2022, the median follow­up was 34.7 months in both cohorts. In the premenopausal cohort, median PFS was 27.6 months in the ribociclib arm (n = 79) versus 14.7 months in the placebo arm (n = 77) (hazard ratio 0.67 [95% CI: 0.45, 1.01]). In the postmenopausal cohort, median PFS was not reached in the ribociclib arm versus 18.5 months in the placebo arm (n = 77 in each arm) (hazard ratio 0.40 [95% CI: 0.26, 0.62]). Data also suggested improvements in secondary efficacy endpoints, although OS data were not mature. The safety profile in this population was consistent with that in global studies. CONCLUSIONS: These data demonstrate a favorable benefit­risk profile for ribociclib + ET in Chinese patients.


Sujet(s)
Aminopyridines , Protocoles de polychimiothérapie antinéoplasique , Tumeurs du sein , Létrozole , Post-ménopause , Purines , Récepteur ErbB-2 , Récepteurs des oestrogènes , Humains , Aminopyridines/administration et posologie , Aminopyridines/usage thérapeutique , Aminopyridines/effets indésirables , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/mortalité , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Purines/administration et posologie , Purines/effets indésirables , Adulte d'âge moyen , Récepteur ErbB-2/métabolisme , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Récepteurs des oestrogènes/métabolisme , Létrozole/administration et posologie , Létrozole/usage thérapeutique , Adulte , Chine , Sujet âgé , Récepteurs à la progestérone/métabolisme , Préménopause , Survie sans progression , Goséréline/administration et posologie , Goséréline/usage thérapeutique , Inhibiteurs de l'aromatase/administration et posologie , Inhibiteurs de l'aromatase/usage thérapeutique , Peuples d'Asie de l'Est
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE