Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Exp Hematol Oncol ; 13(1): 41, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622749

ABSTRACT

Despite major therapeutic advances for two decades, including the most recently approved anti-HER2 drugs, brain metastatic localizations remain the major cause of death for women with metastatic HER2 breast cancer. The main reason is the limited drug passage of the blood-brain barrier after intravenous injection and the significant efflux of drugs, including monoclocal antibodies, after administration into the cerebrospinal fluid. We hypothesized that this efflux was linked to the presence of a FcRn receptor in the blood-brain barrier. To overcome this efflux, we engineered two Fab fragments of trastuzumab, an anti-HER2 monoclonal antibody, and did a thorough preclinical development for therapeutic translational purpose. We demonstrated the safety and equal efficacy of the Fabs with trastuzumab in vitro, and in vivo using a patient-derived xenograft model of HER2 overexpressing breast cancer. For the pharmacokinetic studies of intra-cerebrospinal fluid administration, we implemented original rat models with catheter implanted into the cisterna magna. After intraventricular administration in rats, we demonstrated that the brain-to-blood efflux of Fab was up to 10 times lower than for trastuzumab, associated with a two-fold higher brain penetration compared to trastuzumab. This Fab, capable of significantly reducing brain-to-blood efflux and enhancing brain penetration after intra-cerebrospinal fluid injection, could thus be a new and original effective drug in the treatment of HER2 breast cancer brain metastases, which will be demonstrated by a phase I clinical trial dedicated to women in resort situations.

3.
Bull Cancer ; 110(10): 1041-1050, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37659907

ABSTRACT

INTRODUCTION: In October 2020, the French Health Authority granted early access outside of the clinical trial setting for dostarlimab, a programmed death-1 inhibitor. Dostarlimab was approved by the European Medicines Agency (in April 2021) as monotherapy for patients with post-platinum mismatch repair deficient/microsatellite instability-high advanced/recurrent endometrial cancer, based on the results of the GARNET trial (NCT02715284). METHODS: This was a real-world descriptive analysis of patients granted cohort temporary authorization of use to receive dostarlimab between November 2020 and June 2021. Physicians could complete follow-up forms at each treatment cycle to provide clinical information, safety, and efficacy data. Safety and disease progression data were also captured through pharmacovigilance reports. RESULTS: Of 95 temporary authorization of use requests made by 80 oncologists in 59 French hospitals, 87 patients were eligible, and 80 received≥1 dose of dostarlimab. Based on treatment response assessments received (n=43), the mean (standard deviation) time from treatment initiation to response evaluation was 11 (6) weeks. The disease control rate (complete plus partial responses plus stable disease rates) was 56% (n=24/43), and the overall response rate was 35% (n=15/43); both consistent with those reported in the GARNET trial. No new safety signals were reported. DISCUSSION: The enrolment of 80 patients in an 8-month period highlights the need for access to novel treatment regimens in France for these patients post-platinum. Prospective randomized studies are ongoing to assess the efficacy and safety of dostarlimab and other checkpoint inhibitors as first-line treatment in patients with endometrial cancer.


Subject(s)
Endometrial Neoplasms , Platinum , Female , Humans , Antibodies, Monoclonal, Humanized/therapeutic use , Chronic Disease , DNA Mismatch Repair , Endometrial Neoplasms/drug therapy , Microsatellite Instability , Prospective Studies , Clinical Trials as Topic
4.
Cancers (Basel) ; 15(9)2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37173986

ABSTRACT

The number of publications on nanomedicine in oncology has been exponential over the last ten years, going from 640 publications in 2012 to 2487 publications in 2022, reflecting the growing interest and potential of these new technologies [...].

5.
Exp Hematol Oncol ; 12(1): 46, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37189165

ABSTRACT

BACKGROUND: Clear-cell renal cell carcinomas (ccRCCs) are malignant tumors with high metastatic potential and resistance to treatments occurs almost constantly. Compared to primary tumors, there are still limited genomic data that has been obtained from metastatic samples. METHODS: We aimed to characterize metastatic ccRCC by way of whole-genome analyses of metastatic formalin-fixed samples, using OncoScan® technology. We identified a frequent, unexpected pL1575P NOTCH1 mutation which we set out to characterize for translational purposes. We thus implemented patient-derived xenografts from metastatic samples of human ccRCC to explore its clinical significance. RESULTS: We showed that pL1575P NOTCH1 mutation was an activating mutation, leading to the expression of NOTCH1-intracellular domain-active fragments in both cancer cells and tumor endothelial cells, suggesting a trans-differentiation of cancer cells into tumor micro-vessels. We demonstrated that this mutation could be used as a predictive biomarker of response to CB-103, a specific NOTCH1-intracellular domain inhibitor. One striking result was the considerable anti-angiogenic effect, coherent with the presence of NOTCH1 mutation in tumor micro-vessels. CONCLUSIONS: We identified a frequent, unexpected pL1575P_c4724T_C NOTCH1 mutation as a new biomarker for ccRCC metastases, predictive of response to the CB103 NOTCH1-intracellular domain inhibitor.

6.
Cancers (Basel) ; 15(6)2023 Mar 12.
Article in English | MEDLINE | ID: mdl-36980614

ABSTRACT

Breast cancer brain metastases are a challenging daily practice, and the biological link between gene mutations and metastatic spread to the brain remains to be determined. Here, we performed a meta-analysis on genomic data obtained from primary tumors, extracerebral metastases and brain metastases, to identify gene alterations associated with metastatic processes in the brain. Articles with relevant findings were selected using Medline via PubMed, from January 1999 up to February 2022. A critical review was conducted according to the Preferred Reporting Items for Systematic Review and Meta-analysis statement (PRISMA). Fifty-seven publications were selected for this meta-analysis, including 37,218 patients in all, 11,906 primary tumor samples, 5541 extracerebral metastasis samples, and 1485 brain metastasis samples. We report the overall and sub-group prevalence of gene mutations, including comparisons between primary tumors, extracerebral metastases and brain metastases. In particular, we identified six genes with a higher mutation prevalence in brain metastases than in extracerebral metastases, with a potential role in metastatic processes in the brain: ESR1, ERBB2, EGFR, PTEN, BRCA2 and NOTCH1. We discuss here the therapeutic implications. Our results underline the added value of obtaining biopsies from brain metastases to fully explore their biology, in order to develop personalized treatments.

8.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36834998

ABSTRACT

Anti-HER2 therapies have dramatically improved the prognosis of human epidermal growth factor receptor 2 (HER2)-overexpressing cancers. However, the correlation between the HER2 copy number and the response rate to anti-HER2 remains unclear. Here, following the PRISMA method, we performed a meta-analysis in the neoadjuvant setting in breast cancer to study the association between the HER2 amplification level and the pathological complete response (pCR) to anti-HER2 therapies. Nine articles (four clinical trials, five observational studies) were retrieved after full-text screening, involving 11,238 women with locally advanced breast cancer in the neoadjuvant setting. The median HER2/CEP17 ratio cut-off value was 5.0 ± 5.0 (min-max = 1.0-14.0). For the overall population, the median pCR rate was 48% using the random effect model. The studies were categorized in quartiles as follows: ≤2 (Class 1); 2.1 to 5.0 (Class 2); 5.1 to 7.0 (Class 3); and >7.0 (Class 4). After grouping, the pCR rates were 33%, 49%, 57%, and 79%, respectively. When we excluded the study by Greenwell et al., which accounted for 90% of the patients, using the same quartiles, we still observed an increasing rate of pCR as the HER2/CEP17 ratio increased. This is the first meta-analysis demonstrating the relationship between the HER2 amplification level and the percentage of pCR in the neoadjuvant setting among women with HER2-overexpressing breast cancer, with potential therapeutic applications.


Subject(s)
Breast Neoplasms , Neoadjuvant Therapy , Receptor, ErbB-2 , Female , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Neoadjuvant Therapy/methods , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
10.
Pharmaceutics ; 13(9)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34575525

ABSTRACT

The incidence of brain metastases has been increasing constantly for the last 20 years, because of better control of metastases outside the brain, and the failure of most drugs to cross the blood-brain barrier at relevant pharmacological concentrations. Recent advances in the molecular biology of cancer have led to the identification of numerous molecular alterations, some of them targetable with the development of specific targeted therapies, including tyrosine kinase inhibitors. In this narrative review, we set out to describe the state-of-the-art in the use of tyrosine kinase inhibitors for the treatment of melanoma, lung cancer, and breast cancer brain metastases. We also report preclinical and clinical pharmacological data on brain exposure to tyrosine kinase inhibitors after oral administration and describe the most recent advances liable to facilitate their penetration of the blood-brain barrier at relevant concentrations and limit their physiological efflux.

11.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525427

ABSTRACT

The incidence of brain metastasis has been increasing for 10 years, with poor prognosis, unlike the improvement in survival for extracranial tumor localizations. Since recent advances in molecular biology and the development of specific molecular targets, knowledge of the brain distribution of drugs has become a pharmaceutical challenge. Most anticancer drugs fail to cross the blood-brain barrier. In order to get around this problem and penetrate the brain parenchyma, the use of intrathecal administration has been developed, but the mechanisms governing drug distribution from the cerebrospinal fluid to the brain parenchyma are poorly understood. Thus, in this review we discuss the pharmacokinetics of drugs after intrathecal administration, their penetration of the brain parenchyma and the different systems causing their efflux from the brain to the blood.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain/drug effects , Injections, Spinal/methods , Xenobiotics/pharmacokinetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cerebrospinal Fluid/metabolism , Gene Expression , Glymphatic System/drug effects , Glymphatic System/metabolism , Humans , Neoplasm Metastasis , Permeability , Receptors, Fc/genetics , Receptors, Fc/metabolism , Xenobiotics/metabolism
13.
Aging (Albany NY) ; 12(5): 4230-4246, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32156833

ABSTRACT

BACKGROUND: To develop, validate, and assess the clinical impact of a clinical score to predict a 6-month mortality risk among older cancer patients. RESULTS: The mean age was 81.2 ± 6.1 years (women: 54%, various cancers, metastatic cancer: 45%). The score, namely the GRADE, included two geriatric variables (unintentional weight loss, impaired mobility), two oncological variables (cancer site, cancer extension), and exclusively supportive care. Up to a 14% risk of early death, the decision curves suggest that cancer treatment should be instated. CONCLUSION: We have developed and validated a simple score, easy to implement in daily oncological practice, to predict early death among older cancer patients which could guide oncologists in their treatment decisions. METHODS: 603 outpatients prospectively included in the Physical Frailty in Elderly Cancer patients cohort study. We created a multivariate prediction model by evaluating the strength of the individual components of the Geriatric Assessment regarding risk of death at 6 months. Each component was evaluated by univariate analysis and the significant variables (P ≤ 0.20) were carried on as covariates in the multivariate cox proportion hazard analysis. The beta coefficients from the model were used to build a point-based scoring system. Clinical impact was assessed using decision curves.


Subject(s)
Models, Theoretical , Neoplasms/mortality , Age Factors , Aged , Aged, 80 and over , Female , Geriatric Assessment , Humans , Male , Prognosis , Risk Assessment , Survival Rate
14.
Int J Mol Sci ; 21(1)2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31861465

ABSTRACT

The incidence of brain metastases has increased in the last 10 years. However, the survival of patients with brain metastases remains poor and challenging in daily practice in medical oncology. One of the mechanisms suggested for the persistence of a high incidence of brain metastases is the failure to cross the blood-brain barrier of most chemotherapeutic agents, including the more recent targeted therapies. Therefore, new pharmacological approaches are needed to optimize the efficacy of anticancer drug protocols. In this article, we present recent findings in molecular data on brain metastases. We then discuss published data from pharmacological studies on the crossing of the blood-brain barrier by anticancer agents. We go on to discuss future developments to facilitate drug penetration across the blood-brain barrier for the treatment of brain metastases among cancer patients, using physical methods or physiological transporters.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Animals , Antineoplastic Agents/administration & dosage , Biomarkers , Brain Neoplasms/diagnosis , Cell Membrane Permeability , Drug Administration Routes , Drug Delivery Systems , Drug Development , Humans , Molecular Targeted Therapy
15.
Cancer Chemother Pharmacol ; 83(3): 573-580, 2019 03.
Article in English | MEDLINE | ID: mdl-30610367

ABSTRACT

BACKGROUND: Brain metastases are challenging daily practice in oncology and remain a compartmental problem since most anti-cancer drugs do not cross the blood-brain barrier at relevant pharmacological concentrations. METHODS: In a young woman with HER2-overexpressing breast cancer resistant to standard treatments, at the time of brain metastases progression, a ventricular reservoir was implanted for intrathecal drug injections and detailed pharmacokinetic studies. RESULTS: A first association of intrathecal trastuzumab with intravenous cisplatin was offered to the patient. For trastuzumab, the mean cerebrospinal fluid trough concentration of 53.4 mg/L reached relevant levels, enabling the stabilization of the metastases. Adding intravenous cisplatin was not beneficial, since the cerebrospinal fluid exposure was almost undetectable under 0.08 mg/L. We then offered the patient an intrathecal combination of trastuzumab and methotrexate, because of their in vitro synergic cytotoxicity. The cerebrospinal fluid peak of methotrexate was 1037 µmol/L at 2 h, and the concentrations remained above the theoretical therapeutic concentration. After 2 months of this drug combination, we obtained an excellent response on the brain metastases. CONCLUSION: Our preliminary study supports the interest of a compartmental approach through a direct administration of drugs into the cerebrospinal fluid for the treatment of breast cancer brain metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/therapy , Breast Neoplasms/pathology , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Female , Humans , Infusions, Intravenous , Injections, Spinal , Magnetic Resonance Imaging , Receptor, ErbB-2/metabolism , Trastuzumab/administration & dosage , Trastuzumab/pharmacokinetics , Treatment Outcome
16.
Int J Mol Sci ; 19(12)2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30551640

ABSTRACT

Cancers are heterogeneous at the cell level, and the mechanisms leading to cancer heterogeneity could be clonal evolution or cancer stem cells. Cancer stem cells are resistant to most anti-cancer treatments and could be preferential targets to reverse this resistance, either targeting stemness pathways or cancer stem cell surface markers. Gold nanoparticles have emerged as innovative tools, particularly for photo-thermal therapy since they can be excited by laser to induce hyperthermia. Gold nanoparticles can be functionalized with antibodies to specifically target cancer stem cells. Preclinical studies using photo-thermal therapy have demonstrated the feasibility of targeting chemo-resistant cancer cells to reverse clinical chemoresistance. Here, we review the data linking cancer stem cells and chemoresistance and discuss the way to target them to reverse resistance. We particularly focus on the use of functionalized gold nanoparticles in the treatment of chemo-resistant metastatic cancers.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Gold/therapeutic use , Neoplasms/therapy , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/therapeutic use , Drug Synergism , Female , Gold/pharmacology , Humans , Hyperthermia, Induced , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Neoplastic Stem Cells/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL