Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
Acta Cir Bras ; 39: e391624, 2024.
Article in English | MEDLINE | ID: mdl-38808816

ABSTRACT

PURPOSE: To evaluate the chemotherapeutic activity of temozolomide counter to mammary carcinoma. METHODS: In-vitro anticancer activity has been conducted on MCF7 cells, and mammary carcinoma has been induced in Wistar rats by introduction of 7, 12-Dimethylbenz(a)anthracene (DMBA), which was sustained for 24 weeks. Histopathology, immunohistochemistry, cell proliferation study and apoptosis assay via TUNEL method was conducted to evaluate an antineoplastic activity of temozolomide in rat breast tissue. RESULTS: IC50 value of temozolomide in MCF7 cell has been obtained as 103 µM, which demonstrated an initiation of apoptosis. The temozolomide treatment facilitated cell cycle arrest in G2/M and S phase dose dependently. The treatment with temozolomide suggested decrease of the hyperplastic abrasions and renovation of the typical histological features of mammary tissue. Moreover, temozolomide therapy caused the downregulation of epidermal growth factor receptor, extracellular signal-regulated kinase, and metalloproteinase-1 expression and upstream of p53 and caspase-3 proliferation to indicate an initiation of apoptotic events. CONCLUSIONS: The occurrence of mammary carcinoma has been significantly decreased by activation of apoptotic pathway and abrogation of cellular propagation that allowable for developing a suitable mechanistic pathway of temozolomide in order to facilitate chemotherapeutic approach.


Subject(s)
Antineoplastic Agents, Alkylating , Apoptosis , ErbB Receptors , Rats, Wistar , Temozolomide , Temozolomide/pharmacology , Temozolomide/therapeutic use , Animals , Apoptosis/drug effects , Female , ErbB Receptors/drug effects , ErbB Receptors/antagonists & inhibitors , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Humans , MCF-7 Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Immunohistochemistry , Reproducibility of Results , Rats , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology
2.
Anticancer Res ; 44(5): 1955-1962, 2024 May.
Article in English | MEDLINE | ID: mdl-38677771

ABSTRACT

BACKGROUND/AIM: The epidermal growth factor receptor (EGFR) is over-expressed in several types of cancer, and monoclonal antibody therapy has been the strategy that has shown the best results. This study focused on the construction of a humanized single chain antibody (huscFv) directed against EGFR (HER1). MATERIALS AND METHODS: The CDR grafting method was used to incorporate murine complementarity determining regions (CDRs) of cetuximab into human sequences. A dot blot assay was used to examine the affinity of the huscFv secreted by HEK293T for EGFR. The inhibitory effect on the viability of A549 cells was evaluated using the WST-1 assay. RESULTS: The incorporation of murine CDRs of cetuximab into human sequences increased the degree of humanness by 16.4%. The increase in the humanization of scFv did not affect the affinity for EGFR. Metformin had a dose-dependent effect, with an IC50 of 46 mM, and in combination with huscFv, the cell viability decreased by 45% compared to the 15% demonstrated by huscFv alone. CONCLUSION: The CDR grafting technique is efficient for the humanization of scFv, maintaining its affinity for EGFR and demonstrating its inhibitory effect when combined with metformin in A549 cells.


Subject(s)
Cetuximab , ErbB Receptors , Metformin , Single-Chain Antibodies , Animals , Humans , Mice , A549 Cells/drug effects , Antibodies, Monoclonal, Humanized/pharmacology , Cell Survival/drug effects , Cetuximab/pharmacology , Complementarity Determining Regions/immunology , ErbB Receptors/immunology , ErbB Receptors/antagonists & inhibitors , HEK293 Cells , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/immunology , Metformin/pharmacology , Single-Chain Antibodies/pharmacology , Single-Chain Antibodies/immunology
3.
ChemMedChem ; 19(10): e202300651, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38354370

ABSTRACT

In this research work, a series of 16 quinazoline derivatives bearing ibuprofen and an amino acid were designed as inhibitors of epidermal growth factor receptor tyrosine kinase domain (EGFR-TKD) and cyclooxygenase-2 (COX-2) with the intention of presenting dual action in their biological behavior. The designed compounds were synthesized and assessed for cytotoxicity on epithelial cancer cells lines (AGS, A-431, MCF-7, MDA-MB-231) and epithelial non-tumorigenic cell line (HaCaT). From this evaluation, derivative 6 was observed to exhibit higher cytotoxic potency (IC50) than gefitinib (reference drug) on three cancer cell lines (0.034 µM in A-431, 2.67 µM in MCF-7, and 3.64 µM in AGS) without showing activity on the non-tumorigenic cell line (>100 µM). Furthermore, assessment of EGFR-TKD inhibition by 6 showed a discreet difference compared to gefitinib. Additionally, 6 was used to conduct an in vivo anti-inflammatory assay using the 12-O-tetradecanoylphorbol-3-acetate (TPA) method, and it was shown to be 5 times more potent than ibuprofen. Molecular dynamics studies of EGFR-TKD revealed interactions between compound 6 and M793. On the other hand, one significant interaction was observed for COX-2, involving S531. The RMSD graph indicated that the ligand remained stable in 50 ns.


Subject(s)
Amino Acids , Antineoplastic Agents , Cyclooxygenase 2 , Drug Screening Assays, Antitumor , ErbB Receptors , Ibuprofen , Quinazolines , Ibuprofen/pharmacology , Ibuprofen/chemistry , Ibuprofen/chemical synthesis , Humans , Quinazolines/pharmacology , Quinazolines/chemistry , Quinazolines/chemical synthesis , Cyclooxygenase 2/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Amino Acids/chemistry , Amino Acids/pharmacology , Amino Acids/chemical synthesis , Molecular Structure , Cell Line, Tumor , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Tetradecanoylphorbol Acetate/pharmacology , Cell Proliferation/drug effects , Animals , Dose-Response Relationship, Drug , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Molecular Docking Simulation , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis , Cell Survival/drug effects
4.
An Bras Dermatol ; 98(4): 429-439, 2023.
Article in English | MEDLINE | ID: mdl-36990917

ABSTRACT

The frequency of the use of drugs that act on the epidermal growth factor receptor (EGFR) is increasing, with the consequent onset of cutaneous toxicity, specifically acneiform eruption. The authors extensively review the topic, focusing on describing how these drugs can affect the skin and its appendages, that is, the pathophysiology that encompasses the cutaneous toxicity related to the use of EGFR inhibitors. In addition, it was possible to list the risk factors that may be associated with adverse effects of these drugs. Based on this recent knowledge, the authors expect to aid in the management of patients who are more vulnerable to toxicity, reduce morbidities, and improve the quality of life of patients undergoing treatment with EGFR inhibitors. Other issues related to the toxicity of EGFR inhibitors, such as the clinical aspects of the acneiform eruption grades, and other different types of cutaneous and mucosal reactions, are also included in the article.


Subject(s)
Acneiform Eruptions , Antineoplastic Agents , Drug-Related Side Effects and Adverse Reactions , ErbB Receptors , Humans , Acneiform Eruptions/chemically induced , Acneiform Eruptions/drug therapy , Antineoplastic Agents/adverse effects , Biomarkers , ErbB Receptors/antagonists & inhibitors , Quality of Life , Risk Factors
5.
Québec; INESSS; 6 oct. 2022.
Non-conventional in English | BRISA/RedTESA | ID: biblio-1510544

ABSTRACT

LE POTENTIEL DES DONNÉES CLINICO-ADMINISTRATIVES POUR AMÉLIORER LES PRATIQUES EN CANCÉROLOGIE: L'INESSS a entrepris un vaste chantier en trois volets pour explorer le potentiel des données clinico-administratives comme levier d'amélioration des pratiques en cancérologie. Les présents travaux font référence au deuxième volet, et portent sur l'évaluation de la valeur des thérapies innovantes en contexte réel québécois. ILS ONT POUR OBJECTIFS: de brosser un portrait de l'utilisation des inhibiteurs de la tyrosine kinase du récepteur du facteur de croissance épidermique (EGFR-ITK) au Québec; d'estimer la survie globale des personnes qui reçoivent un EGFR-ITK pour le traitement du cancer du poumon, dans trois indications, et de déterminer si ces résultats sont similaires à ceux rapportés dans les études publiées; d'apprécier l'état des pratiques au Québec et de déterminer des pistes d'amélioration. LES DONNÉES CLINICO-ADMINISTRATIVES: UTILES POUR L'ÉVALUATION DES THÉRAPIES INNOVANTES: Les travaux réalisés montrent qu'il est possible de brosser un portrait de l'utilisation de certaines thérapies innovantes et d'estimer leur valeur en contexte réel québécois à l'aide des banques de données clinico-administratives. Cependant, il convient de retenir les deux points suivants : certains renseignements utiles ne sont pas disponibles, limitant ainsi les types de thérapies et les indications qui peuvent être évaluées, de même que les méthodes d'évaluation qui peuvent être appliquées; un délai relativement important peut être nécessaire pour obtenir des résultats d'une précision raisonnable sur la survie globale médiane. Pour ces travaux, une cohorte globale constituée des personnes qui ont reçu un EGFR-ITK entre le 1er avril 2001 et le 31 mars 2019 a d'abord été créée, puis trois cohortes correspondant aux indications d'intérêt ont été constituées. ACCÉLÉRER L'ACCÈS AUX NOUVELLES THÉRAPIES: Les analyses suggèrent qu'environ cinq ans ont été nécessaires avant la pleine intégration des EGFR-ITK dans la pratique québécoise. Certains cliniciens semblent toutefois avoir été particulièrement proactifs pour intégrer l'osimertinib à leur pratique. Quelques pistes devraient être envisagées pour accélérer l'accès aux nouvelles thérapies, plus particulièrement lorsque les critères d'admissibilité s'appuient sur de nouveaux paradigmes : prévoir des corridors de services pour faciliter l'accès aux tests compagnons dans toutes les régions du Québec, de même que des ressources pour livrer les résultats dans un délai approprié; améliorer la diffusion de l'information sur la disponibilité des nouvelles thérapies auprès des gestionnaires et des cliniciens; favoriser la diffusion de cette même information auprès des patients et associations de patients. ACCÉLÉRER L'ACCÈS AUX NOUVELLES THÉRAPIES: Les analyses suggèrent qu'environ cinq ans ont été nécessaires avant la pleine intégration des EGFR-ITK dans la pratique québécoise. Certains cliniciens semblent toutefois avoir été particulièrement proactifs pour intégrer l'osimertinib à leur pratique. Quelques pistes devraient être envisagées pour accélérer l'accès aux nouvelles thérapies, plus particulièrement lorsque les critères d'admissibilité s'appuient sur de nouveaux paradigmes : prévoir des corridors de services pour faciliter l'accès aux tests compagnons dans toutes les régions du Québec, de même que des ressources pour livrer les résultats dans un délai approprié; améliorer la diffusion de l'information sur la disponibilité des nouvelles thérapies auprès des gestionnaires et des cliniciens; favoriser la diffusion de cette même information auprès des patients et associations de patients. DES PERSPECTIVES PROMETTEUSES: Le troisième volet de ce chantier exploratoire est en cours. À terme, les résultats les plus utiles des trois volets de ce projet pourront être mis à jour sur une base régulière, d'autres questions visant à améliorer les soins pourront être abordées et les méthodes développées pour l'étude du cancer du poumon pourront éventuellement être adaptées à d'autres types de cancer.


Subject(s)
Humans , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Health Evaluation , Cost-Benefit Analysis/trends
6.
J Mol Model ; 27(9): 239, 2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34363097

ABSTRACT

Protein kinases (in this case, HER-2 and EGFR) are involved in cancer-related diseases. Some reports have shown unique CoMFA models using the sum of activities expressed as pIC50 (-log IC50), as the classical CoMFA technique would not be the best strategy to construct models for multitarget therapy considering that the molecular alignment will not be the same for different targets. An alternative for this problem is the use of Topomer-CoMFA, a variation of CoMFA, which does not require the alignment step in the generation of 3D models. In this study, we propose the combined use of the sum of activities and Topomer-CoMFA for the construction of a unique dual 3D model considering the inhibitory activities against EGFR and HER-2. For this, 88 compounds from the literature were divided into two groups: training (71) and test (17) sets. The biological activity of each compound, expressed as IC50 for EGFR and HER-2, was transformed into pIC50, summed, and used as the dependent variable in the Topomer-CoMFA analyses. The obtained model was considered statistically robust in the prediction of the dual activity of new compounds. Finally, based on the obtained model, we proposed structural modifications to some of the compounds used to improve the biological data. From the 3D model, we suggested new derivative compounds with improved biological activity for both targets. Therefore, the combination of the techniques proposed in this study proves to be a good strategy to construct better statistical models that can predict biological activities in multitarget systems.


Subject(s)
Models, Molecular , Protein Kinase Inhibitors/chemistry , Receptor, ErbB-2 , Software , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , Humans , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/chemistry
7.
J Mol Model ; 27(7): 211, 2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34173883

ABSTRACT

Chalcones (E)-1,3-diphenyl-2-propene-1-ones, a class of biosynthetic precursor molecules of flavonoids, have a wide variety of biological applications. Besides the natural products, many synthetic derivatives and analogs became an object of continued interest in academia and industry. In this work, a synthesis and an extensive structural study were performed on a sulfonamide chalcone 1-Benzenesulfonyl-3-(4-bromobenzylidene)-2-(2-chlorophenyl)-2,3-dihydro-1H-quinolin-4-one with potential antineoplastic application. In addition, in silico experiments have shown that the sulfonamide chalcone fits well in the ligand-binding site of EGFR with seven µ-alkyl binding energy interactions on the ligand-binding site. Finally, the kinetic stability and the pharmacophoric analysis for EGFR indicated the necessary spatial characteristics for potential activity of sulfonamide chalcone as an antagonist.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Molecular Docking Simulation , Antineoplastic Agents/pharmacology , Structure-Activity Relationship
8.
Cancer Biomark ; 32(2): 147-160, 2021.
Article in English | MEDLINE | ID: mdl-34151841

ABSTRACT

BACKGROUND: Penile cancer (PeCa) is a rare disease, but its incidence has increased worldwide, mostly in HPV+ patients. Nevertheless, there is still no targeted treatment for this carcinoma. OBJECTIVE: To predict the main signaling pathways involved in penile tumorigenesis and its potential drug targets. METHODS: Genome-wide copy number profiling was performed in 28 PeCa. Integration analysis of CNAs and miRNAs and mRNA targets was performed by DIANA-TarBase v.8. The potential impact of the miRNAs/target genes on biological pathways was assessed by DIANA-miRPath v.3.0. For each miRNA, KEGG pathways were generated based on the tarbase and microT-CDS algorithms. Pharmaco-miR was used to identify associations between miRNAs and their target genes to predict druggable targets. RESULTS: 269 miRNAs and 2,395 genes were mapped in cytobands with CNAs. The comparison of the miRNAs mapped at these cytobands and the miRNAs that were predicted to regulate the genes also mapped in these regions, resulted in a set of common 35 miRNAs and 292 genes. Enrichment pathway revealed their involvement in five top signaling pathways. EGFR and COX2 were identified as potential druggable targets. CONCLUSION: Our data indicate the potential use of EGFR and COX2 inhibitors as a target treatment for PeCa patients.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/genetics , Papillomavirus Infections/genetics , Penile Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Carcinogenesis/genetics , Carcinoma/drug therapy , Carcinoma/pathology , Carcinoma/virology , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , DNA Copy Number Variations , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/metabolism , Middle Aged , Molecular Targeted Therapy/methods , Papillomavirus Infections/drug therapy , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Penile Neoplasms/drug therapy , Penile Neoplasms/pathology , Penile Neoplasms/virology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , RNA, Messenger/metabolism , Signal Transduction/genetics
9.
BMC Cancer ; 21(1): 652, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34074257

ABSTRACT

BACKGROUND: Even though targeted therapies are available for cancers expressing oncogenic epidermal growth receptor (EGFR) and (or) human EGFR2 (HER2), acquired or intrinsic resistance often confounds therapy success. Common mechanisms of therapy resistance involve activating receptor point mutations and (or) upregulation of signaling downstream of EGFR/HER2 to Akt and (or) mitogen activated protein kinase (MAPK) pathways. However, additional pathways of resistance may exist thus, confounding successful therapy. METHODS: To determine novel mechanisms of EGFR/HER2 therapy resistance in breast cancer, gefitinib or lapatinib resistant variants were created from SKBR3 breast cancer cells. Syngenic therapy sensitive and resistant SKBR3 variants were characterized for mechanisms of resistance by mammosphere assays, viability assays, and western blotting for total and phospho proteins. RESULTS: Gefitinib and lapatinib treatments reduced mammosphere formation in the sensitive cells, but not in the therapy resistant variants, indicating enhanced mesenchymal and cancer stem cell-like characteristics in therapy resistant cells. The therapy resistant variants did not show significant changes in known therapy resistant pathways of AKT and MAPK activities downstream of EGFR/HER2. However, these cells exhibited elevated expression and activation of the small GTPase Rac, which is a pivotal intermediate of GFR signaling in EMT and metastasis. Therefore, the potential of the Rac inhibitors EHop-016 and MBQ-167 to overcome therapy resistance was tested, and found to inhibit viability and induce apoptosis of therapy resistant cells. CONCLUSIONS: Rac inhibition may represent a viable strategy for treatment of EGFR/HER2 targeted therapy resistant breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , rac GTP-Binding Proteins/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carbazoles/pharmacology , Carbazoles/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Gain of Function Mutation , Gefitinib/pharmacology , Gefitinib/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Point Mutation , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Spheroids, Cellular , Up-Regulation
10.
Clin Transl Oncol ; 23(8): 1549-1560, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33474678

ABSTRACT

OBJECTIVES: Epithelial growth factor receptor (EGFR), as a malignancy marker, is overly expressed in multiple solid tumors including colorectal neoplasms, one of the most prevalent malignancies worldwide. The main objective of this study is to enhance the efficacy of anti-tumor therapy targeting EGFR by constructing a novel EGFR-specific immunotoxin (C-CUS245C) based on Cetuximab and recombinant Cucurmosin (CUS245C). METHODS: E. coli BL21 (DE3) PlysS (E. coli) was used to express CUS245C with a cysteine residue inserting to the C-terminus of Cucurmosin. Then immobilized metal ion affinity chromatography (IMAC) was used to purify CUS245C. The chemical conjugation method was used for the preparation of C-CUS245C. Then dialysis and IMAC were used to purify C-CUS245C. Western blot as well as SDS-PAGE was carried out to characterize the formation of C-CUS245C. At last the anti-colorectal cancer activity of C-CUS245C was investigated in vitro and in vivo. RESULTS: CUS245C with high purity could be obtained from the prokaryotic system. C-CUS245C was successfully constructed and highly purified. The cytotoxicity assays in vitro showed a significant proliferation inhibition of C-CUS245C on EGFR-positive cells for 120 h with IC50 values less than 0.1 pM. Besides, the anti-tumor efficacy of C-CUS245C was remarkably more potent than that of Cetuximab, CUS245C, and C + CUS245C (P < 0.001). Whereas the cytotoxicity of C-CUS245C could hardly be detected on EGFR-null cell line. Our results also showed that C-CUS245C had efficacy of anti-colorectal cancer in mouse xenograft model, indicating the therapeutic potential of C-CUS245C for the targeted therapy of colorectal neoplasms. CONCLUSIONS: C-CUS245C exhibits potent and EGFR-specific cytotoxicity. Insertional mutagenesis technique is worthy to be adopted in the preparation of immunotoxin. Immunotoxin can be highly purified through dialysis followed by IMAC.


Subject(s)
Cetuximab/therapeutic use , Colorectal Neoplasms/therapy , Immunotoxins/therapeutic use , Molecular Targeted Therapy/methods , Plant Proteins/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab/pharmacology , Chromatography, Affinity/methods , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Escherichia coli/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/therapeutic use , Immunotoxins/chemistry , Immunotoxins/isolation & purification , Immunotoxins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mutagenesis, Insertional/methods , Plant Proteins/chemistry , Plant Proteins/metabolism , Time Factors , Xenograft Model Antitumor Assays
11.
JCO Precis Oncol ; 5: 839-848, 2021 11.
Article in English | MEDLINE | ID: mdl-34994616

ABSTRACT

PURPOSE: BIM activation is essential for epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI)-triggered apoptosis in EGFR-mutant non-small-cell lung cancer (NSCLC). A deletion in the intron two of the BIM gene results in generation of alternatively spliced isoforms that impairs their apoptotic response to TKIs, conferring the NSCLC cells intrinsic resistance to these medications. Patients with both alterations have poor clinical evolution. The current study aimed to investigate the clinical efficacy and tolerability of EGFR-TKIs plus bevacizumab (Bev) versus EGFR-TKIs alone as first-line treatment in advanced NSCLC patients with EGFR mutations and BIM deletions (BIMdel). MATERIALS AND METHODS: A retrospective analysis was conducted. BIMdel was detected using polymerase chain reaction analysis and direct sequencing of DNA. BIM protein expression was investigated by immunohistochemistry, and BIM mRNA levels by reverse transcriptase-polymerase chain reaction. Clinical characteristics, overall survival, progression-free survival (PFS), overall response rate (ORR), and treatment-related adverse events were compared between both groups. RESULTS: Thirty-three patients were included; 15 received EGFR-TKIs, and 18 received EGFR-TKIs plus Bev. The median age was 63 years, with a majority of recruited female patients. All included individuals had an Eastern Cooperative Oncology Group performance score of 2 or less. The addition of Bev resulted in a significantly higher ORR (94.4% v 40%, P > .001). Median PFS was longer with the use of the combination therapy (11.12 v 7.87 months; P = .001). Median overall survival tended to be longer in the EGFR-TKIs plus Bev (30.9 v 25.4 months; P = .06) but failed to reach statistical significance. Response in terms of both partial and complete as well as overall favorably affected PFS. CONCLUSION: EGFR-TKIs plus Bev conferred a significantly higher ORR and PFS in advanced NSCLC patients with EGFR mutation and BIMdel. Further prospective studies are needed to validate these findings.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Bcl-2-Like Protein 11/genetics , Bevacizumab/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Drug Combinations , ErbB Receptors/antagonists & inhibitors , Female , Gene Deletion , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Neoplasm Staging , Polymorphism, Genetic , Retrospective Studies , Young Adult
12.
Clin Transl Oncol ; 23(4): 827-839, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32789773

ABSTRACT

OBJECTIVE: To provide guidance for the management of RAS wild-type (wt) metastatic colorectal cancer (mCRC) in daily practice. METHODS: Nominal group and Delphi techniques were used. A steering committee of seven experts analyzed the current management of RAS wt mCRC, through which they identified controversies, critically analyzed the available evidence, and formulated several guiding statements for clinicians. Subsequently, a group of 30 experts (the expert panel) was selected to test agreement with the statements, through two Delphi rounds. The following response categories were established in both rounds: 1 = totally agree, 2 = basically agree, 3 = basically disagree, 4 = totally disagree. Agreement was defined if ≥ 75% of answers were in categories 1 and 2 (consensus with the agreement) or 3 and 4 (consensus with the disagreement). RESULTS: Overall, 71 statements were proposed, which incorporated the following areas: (1) overarching principles; (2) tumor location; (3) triplets; (4) maintenance; (5) second-line and beyond treatments; (6) Rechallenge and liquid biopsy. After the two Delphi rounds, only six statements maintained a lack of clear consensus. CONCLUSIONS: This document aims to describe the expert's attitude when dealing with several common clinical questions regarding patients with RAS wt mCRC.


Subject(s)
Advisory Committees , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Consensus , Delphi Technique , Genes, ras/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colon, Ascending/pathology , Colon, Transverse/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , Fluorouracil/therapeutic use , Genotype , Humans , Leucovorin/therapeutic use , Liquid Biopsy , Maintenance Chemotherapy/methods , Organoplatinum Compounds/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Retreatment
13.
Hepatology ; 73(4): 1494-1508, 2021 04.
Article in English | MEDLINE | ID: mdl-32602149

ABSTRACT

BACKGROUND AND AIMS: The liver is a highly regenerative organ, but its regenerative capacity is compromised in severe liver injury settings. In chronic liver diseases, the number of liver progenitor cells (LPCs) correlates proportionally to disease severity, implying that their inefficient differentiation into hepatocytes exacerbates the disease. Moreover, LPCs secrete proinflammatory cytokines; thus, their prolonged presence worsens inflammation and induces fibrosis. Promoting LPC-to-hepatocyte differentiation in patients with advanced liver disease, for whom liver transplantation is currently the only therapeutic option, may be a feasible clinical approach because such promotion generates more functional hepatocytes and concomitantly reduces inflammation and fibrosis. APPROACH AND RESULTS: Here, using zebrafish models of LPC-mediated liver regeneration, we present a proof of principle of such therapeutics by demonstrating a role for the epidermal growth factor receptor (EGFR) signaling pathway in differentiation of LPCs into hepatocytes. We found that suppression of EGFR signaling promoted LPC-to-hepatocyte differentiation through the mitogen-activated ERK kinase (MEK)-extracellular signal-regulated kinase (ERK)-sex-determining region Y-box 9 (SOX9) cascade. Pharmacological inhibition of EGFR or MEK/ERK promoted LPC-to-hepatocyte differentiation as well as genetic suppression of the EGFR-ERK-SOX9 axis. Moreover, Sox9b overexpression in LPCs blocked their differentiation into hepatocytes. In the zebrafish liver injury model, both hepatocytes and biliary epithelial cells contributed to LPCs. EGFR inhibition promoted the differentiation of LPCs regardless of their origin. Notably, short-term treatment with EGFR inhibitors resulted in better liver recovery over the long term. CONCLUSIONS: The EGFR-ERK-SOX9 axis suppresses LPC-to-hepatocyte differentiation during LPC-mediated liver regeneration. We suggest EGFR inhibitors as a proregenerative therapeutic drug for patients with advanced liver disease.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Regeneration/drug effects , MAP Kinase Signaling System/drug effects , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Butadienes/pharmacology , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hepatocytes/cytology , Nitriles/pharmacology , Quinazolines/pharmacology , Stem Cells/cytology , Tyrphostins/pharmacology
14.
BMC Cancer ; 20(1): 829, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32873256

ABSTRACT

BACKGROUND: Tyrosine-kinase inhibitors (TKIs) have become the cornerstone treatment of patients with non-small cell lung cancer that harbor oncogenic EGFR mutations. The counterpart of these drugs is the financial burden that they impose, which often creates a barrier for accessing treatment in developing countries. The aim if the present study was to compare the cost-effectiveness of three different first and second generation TKIs. METHODS: We designed a retrospective cost-effectiveness analysis of three different TKIs (afatinib, erlotinib, and gefitinib) administered as first-line therapy for patients with NSCLC that harbor EGFR mutations. RESULTS: We included 99 patients with the following TKI treatment; 40 treated with afatinib, 33 with gefitinib, and 26 with erlotinib. Median PFS was not significantly different between treatment groups; 15.4 months (95% CI 9.3-19.5) for afatinib; 9.0 months (95% CI 6.3- NA) for erlotinib; and 10.0 months (95% CI 7.46-14.6) for gefitinib. Overall survival was also similar between groups: 29.1 months (95% CI 25.4-NA) for afatinib; 27.1 months (95% CI 17.1- NA) for erlotinib; and 23.7 months (95% CI 18.6-NA) for gefitinib. There was a statistically significant difference between the mean TKIs costs; being afatinib the most expensive treatment. This difference was observed in the daily cost of treatment (p < 0.01), as well as the total cost of treatment (p = 0.00095). Cost-effectiveness analysis determined that afatinib was a better cost-effective option when compared with first-generation TKIs (erlotinib and gefitinib). CONCLUSION: In our population, erlotinib, afatinib, and gefitinib were statistically equally effective in terms of OS and PFS for the treatment of patients with advanced EGFR-mutated NSCLC population. Owing to its marginally increased PFS and OS, the cost-effectiveness analysis determined that afatinib was a slightly better cost-effective option when compared with first-generation TKIs (erlotinib and gefitinib).


Subject(s)
Afatinib/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cost-Benefit Analysis/methods , Erlotinib Hydrochloride/administration & dosage , Gefitinib/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/administration & dosage , Adult , Afatinib/economics , Aged , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride/economics , Female , Gefitinib/economics , Humans , Male , Middle Aged , Progression-Free Survival , Protein Kinase Inhibitors/economics , Retrospective Studies
15.
Mol Biol Rep ; 47(9): 6817-6828, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32862352

ABSTRACT

A member of the Trk family of neurotrophin receptors, tropomyosin receptor kinase B (TrkB, encoded by the NTRK2 gene) is an increasingly important target in various cancer types, including glioblastoma (GBM). EGFR is among the most frequently altered oncogenes in GBM, and EGFR inhibition has been tested as an experimental therapy. Functional interactions between EGFR and TrkB have been demonstrated. In the present study, we investigated the role of TrkB and EGFR, and their interactions, in GBM. Analyses of NTRK2 and EGFR gene expression from The Cancer Genome Atlas (TCGA) datasets showed an increase in NTRK2 expression in the proneural subtype of GBM, and a strong correlation between NTRK2 and EGFR expression in glioma CpG island methylator phenotype (G-CIMP+) samples. We showed that when TrkB and EGFR inhibitors were combined, the inhibitory effect on A172 human GBM cells was more pronounced than when either inhibitor was given alone. When U87MG GBM cells were xenografted into the flank of nude mice, tumor growth was delayed by treatment with TrkB and EGFR inhibitors, given alone or combined, only at specific time points. Intracranial GBM growth in mice was not significantly affected by drug treatments. Our findings indicate that correlations between NTRK2 and EGFR expression occur in specific GBM subgroups. Also, our results using cultured cells suggest for the first time the potential of combining TrkB and EGFR inhibition for the treatment of GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , Glioblastoma/metabolism , Membrane Glycoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, trkB/metabolism , Animals , Azepines/pharmacology , Benzamides/pharmacology , Brain Neoplasms/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Synergism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/genetics , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Grading , Quinazolines/pharmacology , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/genetics , Tyrphostins/pharmacology , Xenograft Model Antitumor Assays
16.
Photochem Photobiol ; 96(3): 684-691, 2020 05.
Article in English | MEDLINE | ID: mdl-32119123

ABSTRACT

Peritoneal carcinomatosis (PC) can occur as an advanced consequence of multiple primary malignancies. Surgical resection, radiation or systemic interventions alone have proven inadequate for this aggressive cancer presentation, since PC still has a poor survival profile. Photodynamic therapy (PDT), in which photosensitive drugs are exposed to light to generate cytotoxic reactive oxygen species, may be an ideal treatment for PC because of its ability to deliver treatment to a depth appropriate for peritoneal surface tumors. Additionally, epidermal growth factor receptor (EGFR) signaling plays a variety of roles in cancer progression and survival as well as PDT-mediated cytotoxicity, so EGFR inhibitors may be valuable in enhancing the therapeutic index of intraperitoneal PDT. This study examines escalating doses of benzoporphyrin derivative (BPD)-mediated intraperitoneal PDT combined with the EGFR-inhibitor cetuximab in a canine model. In the presence or absence of small bowel resection (SBR) and cetuximab, we observed a tolerable safety and toxicity profile related to the light dose received. Additionally, our findings that BPD levels are higher in the small bowel compared with other anatomical regions, and that the risk of anastomotic failure decreases at lower light doses will help to inform the design of similar PC treatments in humans.


Subject(s)
Antineoplastic Agents/administration & dosage , Cetuximab/administration & dosage , Disease Models, Animal , Peritoneal Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Cetuximab/pharmacology , Dogs , ErbB Receptors/antagonists & inhibitors , Female , Humans , Male
17.
JAMA Dermatol ; 156(2): 196-200, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31895414

ABSTRACT

Importance: Olmsted syndrome is a rare and disabling genodermatosis for which no successful treatment is currently available. Objective: To evaluate the clinical response to the mammalian target of rapamycin (mTOR) inhibitor sirolimus and/or the epidermal growth factor receptor (EGFR) inhibitor erlotinib among patients with Olmsted syndrome. Design, Setting, and Participants: This case series focused on 4 children with treatment-refractory Olmsted syndrome. These children received treatments (initiated in 2017 and 2018) at the outpatient dermatology clinic at the Children's Hospital of Wisconsin in Milwaukee, Wisconsin; Children's National Hospital in Washington, DC; and Hospital Infantil Pequeno Príncipe, Curitiba in Paraná, Brazil. Exposures: Immunohistochemical analyses for mTOR and EGFR activation were performed on skin biopsy specimens from 2 patients. Oral sirolimus was administered to these 2 patients at a dosage of 0.8 mg/m2 twice daily, titrated to a goal trough whole-blood concentration of 10 to 15 ng/mL. Erlotinib was administered to all 4 patients at a dosage of 2 mg/kg/d. Main Outcomes and Measures: Clinical responses were assessed with visual analog scales for pruritus and pain and/or the Children's Dermatology Life Quality Index. Adverse effects were monitored throughout treatment. Results: Four patients (mean [SD] age, 7 [6] years; 2 boys and 2 girls) were analyzed. Lesional skin immunostaining showed increased phosphorylated ribosomal protein S6 (RPS6) and phosphorylated EGFR staining in the epidermis, indicating enhanced mTOR and EGFR signaling activation. Patients 1 and 2 were initially treated with sirolimus, displaying substantial clinical improvement in erythema and periorificial hyperkeratosis afterward. When switched to erlotinib, these patients showed substantial palmoplantar keratoderma (PPK) improvement. Patients 3 and 4 were treated with erlotinib only and later showed rapid and near complete resolution of PPK and substantial improvement in Children's Dermatology Life Quality Index scores. All 4 patients had sustained improvements in pruritus and pain. No severe adverse effects were reported. Conclusions and Relevance: This study's findings suggest that the EGFR-mTOR cascade may play a substantial role in the pathophysiological process of Olmsted syndrome and may serve as a major therapeutic target. Oral sirolimus and erlotinib may be a promising, life-altering treatment for pediatric patients with Olmsted syndrome.


Subject(s)
Erlotinib Hydrochloride/administration & dosage , Keratoderma, Palmoplantar/drug therapy , Protein Kinase Inhibitors/administration & dosage , Sirolimus/administration & dosage , Adolescent , Brazil , Child , Child, Preschool , ErbB Receptors/antagonists & inhibitors , Female , Humans , Immunosuppressive Agents/administration & dosage , Infant , Keratoderma, Palmoplantar/genetics , Male , Signal Transduction/drug effects , Syndrome , TOR Serine-Threonine Kinases/antagonists & inhibitors , Treatment Outcome
18.
Sci Rep ; 10(1): 1194, 2020 Jan 27.
Article in English | MEDLINE | ID: mdl-31988343

ABSTRACT

Nimotuzumab is a humanized monoclonal antibody against the Epidermal Growth Factor Receptor with a long history of therapeutic use, recognizing an epitope different from the ones targeted by other antibodies against the same antigen. It is also distinguished by much less toxicity resulting in a better safety profile, which has been attributed to its lower affinity compared to these other antibodies. Nevertheless, the ideal affinity window for optimizing the balance between anti-tumor activity and toxic effects has not been determined. In the current work, the paratope of the phage-displayed nimotuzumab Fab fragment was evolved in vitro to obtain affinity-matured variants. Soft-randomization of heavy chain variable region CDRs and phage selection resulted in mutated variants with improved binding ability. Two recombinant antibodies were constructed using these variable regions, which kept the original fine epitope specificity and showed moderate affinity increases against the target (3-4-fold). Such differences were translated into a greatly enhanced inhibitory capacity upon ligand-induced receptor phosphorylation on tumor cells. The new antibodies, named K4 and K5, are valuable tools to explore the role of affinity in nimotuzumab biological properties, and could be used for applications requiring a fine-tuning of the balance between binding to tumor cells and healthy tissues.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Affinity/immunology , Neoplasms/immunology , Antibodies, Monoclonal, Humanized/metabolism , Bacteriophages/genetics , Bacteriophages/immunology , Cell Line, Tumor , Computer Simulation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Genetic Vectors/genetics , Humans , Immunoglobulin Idiotypes/immunology , Immunoglobulin Variable Region/genetics , Neoplasms/pathology , Recombinant Proteins/immunology , Transfection
19.
J Comput Aided Mol Des ; 34(3): 293-303, 2020 03.
Article in English | MEDLINE | ID: mdl-31828486

ABSTRACT

Abnormalities in the expression levels of EGFR/HER2 are found in many different types of human cancer; therefore, the design of dual inhibitors of EGFR/HER2 is a recognized anti-cancer strategy. Some lapatinib derivatives have been previously synthesized by modification at the methylsulfonylethylaminomethylfuryl group and biologically evaluated, demonstrating that the 2i compound shows potent inhibitory activity against EGFR/HER2-overexpressing cancer cells. In the present study, we explored the structural and energetic features that guide the molecular recognition of 2i using various EGFR/HER2 states. Molecular dynamics (MD) simulation with an MMPB(GB)SA approach was used to generate the inactive EGFR/HER2-ligand complexes. Our results corroborate that slight modification of lapatinib contributes to an increase in the affinity of the 2i compound for inactive EGFR/HER2 as compared with lapatinib compound, which is in accordance with experimental results. Comparison with previous results reveals that lapatinib and its derivative bind more strongly to the inactive than the intermediate active-inactive HER2 state. Principal component analysis allowed the observation that coupling of 2i to EGFR/HER2 is linked to a reduction in the conformational mobility, which may also contribute to the improvement in affinity observed for this compound as compared with lapatinib.


Subject(s)
Lapatinib/chemistry , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Receptor, ErbB-2/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Catalytic Domain/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Computational Biology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Lapatinib/therapeutic use , Molecular Dynamics Simulation , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors
20.
Steroids ; 155: 108551, 2020 03.
Article in English | MEDLINE | ID: mdl-31812624

ABSTRACT

Cardiotonic steroids (CTS) are agents traditionally known for their capacity to bind to the Na,K-ATPase (NKA), affecting the ion transport and the contraction of the heart. Natural CTS have been shown to also have effects on cell signaling pathways. With the goal of developing a new CTS derivative, we synthesized a new digoxin derivative, 21-benzylidene digoxin (21-BD). Previously, we have shown that this compound binds to NKA and has cytotoxic actions on cancer, but not on normal cells. Here, we further studied the mechanisms of actions of 21-BD. Working with HeLa cells, we found that 21-BD decreases the basal, as well as the insulin stimulated proliferation. 21-BD reduces phosphorylation of the epidermal growth factor receptor (EGFR) and extracellular-regulated kinase (ERK), which are involved in pathways that stimulate cell proliferation. In addition, 21-BD promotes apoptosis, which is mediated by the translocation of Bax from the cytosol to mitochondria and the release of mitochondrial cytochrome c to the cytosol. 21-BD also activated caspases-8, -9 and -3, and induced the cleavage of poly (ADP-ribose) polymerase-1 (PARP-1). Altogether, these results show that the new compound that we have synthesized exerts cytotoxic actions on HeLa cells by inhibition of cell proliferation and the activation of both the extrinsic and intrinsic apoptotic pathways. These results support the relevance of the cardiotonic steroid scaffold as modulators of cell signaling pathways and potential agents for their use in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Digoxin/analogs & derivatives , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Digoxin/chemistry , Digoxin/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Conformation , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL