Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.884
Filter
1.
BMC Cancer ; 24(1): 526, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664720

ABSTRACT

BACKGROUND: Panel gene sequencing is an established diagnostic tool for precision oncology of solid tumors, but its utility for the treatment of cancers of the digestive system in clinical routine is less well documented. METHODS: We retrospectively identified patients with advanced or metastatic gastrointestinal, pancreaticobiliary or hepatic cancers who received panel gene sequencing at a tertiary university hospital from 2015 to 2022. For these cases, we determined the spectrum of genetic alterations, clinicopathological parameters and treatment courses. Assessment of actionability of genetic alterations was based on the OncoKB database, cancer-specific ESMO treatment guidelines, and recommendations of the local molecular tumor board. RESULTS: In total, 155 patients received panel gene sequencing using either the Oncomine Focus (62 cases), Comprehensive (91 cases) or Childhood Cancer Research Assay (2 cases). The mean age of patients was 61 years (range 24-90) and 37% were female. Most patients suffered from either colorectal cancer (53%) or cholangiocellular carcinoma (19%). 327 genetic alterations were discovered in 123 tumor samples, with an average number of 2.1 alterations per tumor. The most frequently altered genes were TP53, KRAS and PIK3CA. Actionable gene alterations were detected in 13.5-56.8% of tumors, according to ESMO guidelines or the OncoKB database, respectively. Thirteen patients were treated with targeted therapies based on identified molecular alterations, with a median progression-free survival of 8.8 months. CONCLUSIONS: Actionable genetic alterations are frequently detected by panel gene sequencing in patients with advanced cancers of the digestive tract, providing clinical benefit in selected cases. However, for the majority of identified actionable alterations, sufficient clinical evidence for targeted treatments is still lacking.


Subject(s)
Digestive System Neoplasms , Humans , Female , Male , Retrospective Studies , Middle Aged , Aged , Adult , Aged, 80 and over , Young Adult , Digestive System Neoplasms/genetics , Digestive System Neoplasms/pathology , Digestive System Neoplasms/therapy , Mutation , Precision Medicine/methods , Molecular Targeted Therapy/methods , Biomarkers, Tumor/genetics
2.
World J Gastroenterol ; 30(13): 1801-1809, 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38659483

ABSTRACT

Advancements in murine modeling systems for ulcerative colitis have diversified our understanding of the pathophysiological factors involved in disease onset and progression. This has fueled the identification of molecular targets, resulting in a rapidly expanding therapeutic armamentarium. Subsequently, management strategies have evolved from symptomatic resolution to well-defined objective endpoints, including clinical remission, endoscopic remission and mucosal healing. While the incorporation of these assessment modalities has permitted targeted intervention in the context of a natural disease history and the prevention of complications, studies have consistently depicted discrepancies associated with ascertaining disease status through clinical and endoscopic measures. Current recommendations lack consideration of histological healing. The simultaneous achievement of clinical, endoscopic, and histologic remission has not been fully investigated. This has laid the groundwork for a novel therapeutic outcome termed disease clearance (DC). This article summarizes the concept of DC and its current evidence.


Subject(s)
Colitis, Ulcerative , Disease Models, Animal , Intestinal Mucosa , Remission Induction , Colitis, Ulcerative/therapy , Colitis, Ulcerative/diagnosis , Humans , Animals , Intestinal Mucosa/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Remission Induction/methods , Treatment Outcome , Mice , Disease Progression , Molecular Targeted Therapy/methods , Colon/pathology , Colon/drug effects
3.
Sci Prog ; 107(2): 368504241247402, 2024.
Article in English | MEDLINE | ID: mdl-38651330

ABSTRACT

Idiopathic pulmonary fibrosis is a chronic and progressive interstitial lung disease with a poor prognosis. Idiopathic pulmonary fibrosis is characterized by repeated alveolar epithelial damage leading to abnormal repair. The intercellular microenvironment is disturbed, leading to continuous activation of fibroblasts and myofibroblasts, deposition of extracellular matrix, and ultimately fibrosis. Moreover, pulmonary fibrosis was also found as a COVID-19 complication. Currently, two drugs, pirfenidone and nintedanib, are approved for clinical therapy worldwide. However, they can merely slow the disease's progression rather than rescue it. These two drugs have other limitations, such as lack of efficacy, adverse effects, and poor pharmacokinetics. Consequently, a growing number of molecular therapies have been actively developed. Treatment options for IPF are becoming increasingly available. This article reviews the research platform, including cell and animal models involved in molecular therapy studies of idiopathic pulmonary fibrosis as well as the promising therapeutic targets and their development progress during clinical trials. The former includes patient case/control studies, cell models, and animal models. The latter includes transforming growth factor-beta, vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factor, lysophosphatidic acid, interleukin-13, Rho-associated coiled-coil forming protein kinase family, and Janus kinases/signal transducers and activators of transcription pathway. We mainly focused on the therapeutic targets that have not only entered clinical trials but were publicly published with their clinical outcomes. Moreover, this work provides an outlook on some promising targets for further validation of their possibilities to cure the disease.


Subject(s)
Idiopathic Pulmonary Fibrosis , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Humans , Animals , Molecular Targeted Therapy/methods , Pyridones/therapeutic use , Indoles/therapeutic use , Indoles/pharmacology , COVID-19 , Disease Models, Animal
4.
Nat Rev Clin Oncol ; 21(5): 354-369, 2024 May.
Article in English | MEDLINE | ID: mdl-38503878

ABSTRACT

Claudin 18.2, a tight-junction molecule predominantly found in the nonmalignant gastric epithelium, becomes accessible on the tumour cell surface during malignant transformation, thereby providing an appealing target for cancer therapy. Data from two phase III trials testing the anti-claudin 18.2 antibody zolbetuximab have established claudin 18.2-positive advanced-stage gastric cancers as an independent therapeutic subset that derives benefit from the addition of this agent to chemotherapy. This development has substantially increased the percentage of patients eligible for targeted therapy. Furthermore, newer treatments, such as high-affinity monoclonal antibodies, bispecific antibodies, chimeric antigen receptor T cells and antibody-drug conjugates capable of bystander killing effects, have shown considerable promise in patients with claudin 18.2-expressing gastric cancers. This new development has resulted from drug developers moving beyond traditional targets, such as driver gene alterations or growth factors. In this Review, we highlight the biological rationale and explore the clinical activity of therapies that target claudin 18.2 in patients with advanced-stage gastric cancer and explore the potential for expansion of claudin 18.2-targeted therapies to patients with other claudin 18.2-positive solid tumours.


Subject(s)
Claudins , Molecular Targeted Therapy , Stomach Neoplasms , Humans , Claudins/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Molecular Targeted Therapy/methods
5.
Int J Mol Sci ; 25(5)2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38473804

ABSTRACT

Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer, marked by poor outcomes and dismal prognosis. Due to the absence of targetable receptors, chemotherapy still represents the main therapeutic option. Therefore, current research is now focusing on understanding the specific molecular pathways implicated in TNBC, in order to identify novel biomarker signatures and develop targeted therapies able to improve its clinical management. With the aim of identifying novel molecular features characterizing TNBC, elucidating the mechanisms by which these molecular biomarkers are implicated in the tumor development and progression, and assessing the impact on cancerous cells following their inhibition or modulation, we conducted a literature search from the earliest works to December 2023 on PubMed, Scopus, and Web Of Science. A total of 146 studies were selected. The results obtained demonstrated that TNBC is characterized by a heterogeneous molecular profile. Several biomarkers have proven not only to be characteristic of TNBC but also to serve as potential effective therapeutic targets, holding the promise of a new era of personalized treatments able to improve its prognosis. The pre-clinical findings that have emerged from our systematic review set the stage for further investigation in forthcoming clinical trials.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/pathology , Biomarkers, Tumor , Molecular Targeted Therapy/methods
6.
Int J Surg ; 110(4): 2323-2337, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38241327

ABSTRACT

Spindle and kinetochore-associated complex subunit 3 (SKA3) is a microtubule-binding subcomplex of the outer kinetochore, which plays a vital role in proper chromosomal segregation and cell division. Recently, SKA3 have been demonstrated its oncogenic role of tumorigenesis and development in cancers. In this review, the authors comprehensively deciphered SKA3 in human cancer from various aspects, including bibliometrics, pan-cancer analysis, and narrative summary. The authors also provided the top 10 predicted drugs targeting SKA3. The authors proposed that SKA3 was a potential target and brought new therapeutic opportunities for cancer patients.


Subject(s)
Cell Cycle Proteins , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/surgery , Molecular Targeted Therapy/methods , Precision Medicine/methods , Microtubule-Associated Proteins/metabolism
7.
Curr Opin Oncol ; 36(1): 35-43, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37975311

ABSTRACT

PURPOSE OF REVIEW: In this article, we aimed to summarize the results from recent phase III clinical trials that have evaluated the use of immune checkpoint inhibitors (ICIs) in elderly patients with lung cancer. RECENT FINDINGS: Lung cancer is the second most diagnosed malignant tumor and the leading cause of cancer-related deaths worldwide. ICIs have a significant role in the treatment of lung cancer, both as monotherapy and combination therapy prolonged survival benefits. At present, a significant proportion of clinical patients comprise individuals aged 70 years or older. However, the inclusion of elderly patients, particularly in clinical trials involving immunotherapy, remains inadequate, with a limited number of participants from this age group. The lack of evidence regarding the use of ICIs in elderly patients is primarily attributed to the significant underrepresentation of elderly individuals in clinical trials. SUMMARY: In this article, we summarize the results from recent phase III clinical trials that have evaluated the use of ICIs as first-line or second-line monotherapy, in combination with chemotherapy and other immunotherapies in elderly patients with lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Aged , Humans , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Molecular Targeted Therapy/methods , Clinical Trials, Phase III as Topic
8.
Cancer Biomark ; 38(4): 413-424, 2023.
Article in English | MEDLINE | ID: mdl-37980650

ABSTRACT

BACKGROUND: The initiation biomarker-driven trials have revolutionized oncology drug development by challenging the traditional phased approach and introducing basket studies. Notable successes in non-small cell lung cancer (NSCLC) with ALK, ALK/ROS1, and EGFR inhibitors have prompted the need to expand this approach to other cancer sites. OBJECTIVES: This study explores the use of dose response modeling and time-to-event algorithms on the biomarker molecular targeted agent (MTA). By simulating subgroup identification in MTA-related time-to-event data, the study aims to develop statistical methodology supporting biomarker-driven trials in oncology. METHODS: A total of n patients are selected assigned for different doses. A dataset is prepared to mimic the situation on Subgroup Identification of MTA for time to event data analysis. The response is measured through MTA. The MTA value is also measured through ROC. The Markov Chain Monte Carlo (MCMC) techniques are prepared to perform the proposed algorithm. The analysis is carried out with a simulation study. The subset selection is performed through the Threshold Limit Value (TLV) by the Bayesian approach. RESULTS: The MTA is observed with range 12-16. It is expected that there is a marginal level shift of the MTA from pre to post-treatment. The Cox time-varying model can be adopted further as causal-effect relation to establishing the MTA on prolonging the survival duration. The proposed work in the statistical methodology to support the biomarker-driven trial for oncology research. CONCLUSION: This study extends the application of biomarker-driven trials beyond NSCLC, opening possibilities for implementation in other cancer sites. By demonstrating the feasibility and efficacy of utilizing MTA as a biomarker, the research lays the foundation for refining and validating biomarker use in clinical trials. These advancements aim to enhance the precision and effectiveness of cancer treatments, ultimately benefiting patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein-Tyrosine Kinases/therapeutic use , Bayes Theorem , Proto-Oncogene Proteins , Biomarkers/analysis , Receptor Protein-Tyrosine Kinases/therapeutic use , Molecular Targeted Therapy/methods
9.
J Cancer Res Ther ; 19(5): 1206-1211, 2023.
Article in English | MEDLINE | ID: mdl-37787284

ABSTRACT

Introduction: The foundation of oncology treatment as a single modality approach as well as the "multimodality" concept has been studied by statistical evaluation pre, during, and posttreatment to rule out their efficacy, expected prognosis, toxicity reactions, and overall survival for the patient. Such studies have also provided an appreciable amount of data for future custom utility. "Targeted therapy" is a cancer treatment that uses drugs but is different from traditional chemotherapy. It works by targeting cancer-specific genes, proteins, or the tissue environment that contributes to cancer growth and survival. Researchers are developing drugs that target specific molecular changes. The drugs can block or turn off signals that tell cancer cells to grow and divide, keep cells from living longer than usual, and destroy the cancer cells. Aim: The aim of the study is to carry out a systematic review of clinical trials of molecular targeted therapy in the treatment of cancer. Objective: The objective of the study is to evaluate the efficacy of molecular targeted therapy in the treatment of head-and-neck cancers. Materials and Methods: A group of keywords was preselected to search for scientific articles on a web-based database of PubMed. Only completed randomized controlled trials published in the past 5 years in the English language were included with open access. All the selected articles were subjected to the Cochrane bias tool and PRISMA guidelines to extract results. Results: Among 4 studies specifying the progression-free survival (PFS) for comparing the groups treated either using targeted therapy or other modality/placebo, 50% of studies show a slight increase in PFS in the group treated with TT and other 50% show PFS increase in the non-TT group. Thus, insufficient evidence is furnished to provide a statement and acknowledged the expectancy of a disease-free period with or without the use of TT in the treatment of head-and-neck cancer. Conclusion: Considering very little information on enhanced effect and presence of evidence supporting an increased risk of adverse events, the addition of TT to treatment is a question to the dilemma. A systematic review intends advantageous in providing foresight for oncologists concerning patient assessment and evaluation to defend inclination proceeding toward the treatment defined.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/chemically induced
11.
Nature ; 620(7975): 737-745, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37612393

ABSTRACT

The substantial investments in human genetics and genomics made over the past three decades were anticipated to result in many innovative therapies. Here we investigate the extent to which these expectations have been met, excluding cancer treatments. In our search, we identified 40 germline genetic observations that led directly to new targets and subsequently to novel approved therapies for 36 rare and 4 common conditions. The median time between genetic target discovery and drug approval was 25 years. Most of the genetically driven therapies for rare diseases compensate for disease-causing loss-of-function mutations. The therapies approved for common conditions are all inhibitors designed to pharmacologically mimic the natural, disease-protective effects of rare loss-of-function variants. Large biobank-based genetic studies have the power to identify and validate a large number of new drug targets. Genetics can also assist in the clinical development phase of drugs-for example, by selecting individuals who are most likely to respond to investigational therapies. This approach to drug development requires investments into large, diverse cohorts of deeply phenotyped individuals with appropriate consent for genetically assisted trials. A robust framework that facilitates responsible, sustainable benefit sharing will be required to capture the full potential of human genetics and genomics and bring effective and safe innovative therapies to patients quickly.


Subject(s)
Drug Development , Human Genetics , Molecular Targeted Therapy , Humans , Drug Approval/statistics & numerical data , Drug Development/statistics & numerical data , Therapies, Investigational/statistics & numerical data , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/statistics & numerical data , Rare Diseases/genetics , Rare Diseases/therapy , Germ-Line Mutation , Time Factors
12.
Colloids Surf B Biointerfaces ; 229: 113466, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37515959

ABSTRACT

Lung cancer is one of the most aggressive and deadliest health threats. There has been an increasing interest in non-coding RNA (ncRNA) recently, especially in the areas of carcinogenesis and tumour progression. However, ncRNA-directed therapies are still encountering obstacles on their way to the clinic. In the present article, we provide an overview on the potential of targeting ncRNA in the treatment of lung cancer. Then, we discuss the delivery challenges and recent approaches enabling the delivery of ncRNA-directed therapies to the lung cancer cells, where we illuminate some advanced technologies including chemically-modified oligonucleotides, nuclear targeting, and three-dimensional in vitro models. Furthermore, advanced non-viral delivery systems recruiting nanoparticles, biomimetic delivery systems, and extracellular vesicles are also highlighted. Lastly, the challenges limiting the clinical trials on the therapeutic targeting of ncRNAs in lung cancer and future directions to tackle them are explored.


Subject(s)
Lung Neoplasms , RNA, Untranslated , Humans , RNA, Untranslated/genetics , RNA, Untranslated/therapeutic use , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Carcinogenesis , Molecular Targeted Therapy/methods
13.
J Biol Chem ; 299(9): 105111, 2023 09.
Article in English | MEDLINE | ID: mdl-37517694

ABSTRACT

Upon infection by the malaria parasite Plasmodium falciparum, the glycolytic rate of a red blood cell increases up to 100-fold, possibly contributing to lactic acidosis and hypoglycemia in patients with severe malaria. This dramatic increase in glucose uptake and metabolism was correctly predicted by a newly constructed detailed enzyme kinetic model of glucose metabolism in the trophozoite-infected red blood cell. Subsequently, we expanded the model to simulate an infected red blood cell culture, including the different asexual blood-stage forms of the malaria parasite. The model simulations were in good agreement with experimental data, for which the measured parasitic volume was an important parameter. Upon further analysis of the model, we identified glucose transport as a drug target that would specifically affect infected red blood cells, which was confirmed experimentally with inhibitor titrations. This model can be a first step in constructing a whole-body model for glucose metabolism in malaria patients to evaluate the contribution of the parasite's metabolism to the disease state.


Subject(s)
Antimalarials , Erythrocytes , Glycolysis , Malaria, Falciparum , Models, Biological , Molecular Targeted Therapy , Plasmodium falciparum , Humans , Acidosis, Lactic , Antimalarials/pharmacology , Antimalarials/therapeutic use , Antimalarials/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/parasitology , Glucose/metabolism , Glycolysis/drug effects , Hypoglycemia , Kinetics , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Plasmodium falciparum/physiology , Trophozoites/pathogenicity , Trophozoites/physiology , Molecular Targeted Therapy/methods , Parasite Load
14.
Mol Cancer ; 22(1): 105, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37415164

ABSTRACT

Breast cancer is the second leading cause of death for women worldwide. The heterogeneity of this disease presents a big challenge in its therapeutic management. However, recent advances in molecular biology and immunology enable to develop highly targeted therapies for many forms of breast cancer. The primary objective of targeted therapy is to inhibit a specific target/molecule that supports tumor progression. Ak strain transforming, cyclin-dependent kinases, poly (ADP-ribose) polymerase, and different growth factors have emerged as potential therapeutic targets for specific breast cancer subtypes. Many targeted drugs are currently undergoing clinical trials, and some have already received the FDA approval as monotherapy or in combination with other drugs for the treatment of different forms of breast cancer. However, the targeted drugs have yet to achieve therapeutic promise against triple-negative breast cancer (TNBC). In this aspect, immune therapy has come up as a promising therapeutic approach specifically for TNBC patients. Different immunotherapeutic modalities including immune-checkpoint blockade, vaccination, and adoptive cell transfer have been extensively studied in the clinical setting of breast cancer, especially in TNBC patients. The FDA has already approved some immune-checkpoint blockers in combination with chemotherapeutic drugs to treat TNBC and several trials are ongoing. This review provides an overview of clinical developments and recent advancements in targeted therapies and immunotherapies for breast cancer treatment. The successes, challenges, and prospects were critically discussed to portray their profound prospects.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Female , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Immunotherapy/methods , Combined Modality Therapy , Molecular Targeted Therapy/methods
16.
Clin Lymphoma Myeloma Leuk ; 23(10): 705-713, 2023 10.
Article in English | MEDLINE | ID: mdl-37344332

ABSTRACT

Classic Hodgkin lymphoma (HL) is a unique lymphoid malignancy where the malignant cells comprise only 1% to 2% of the total tumor cellularity. Over the past 2 decades, the treatment of HL has evolved drastically based on the advent of novel targeted therapies. Novel agents including programmed death-1 (PD-1) inhibitors, antibody-drug conjugates such as brentuximab vedotin, bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapies have served to shape the management of HL in the frontline as well as the relapsed and refractory (R/R) setting. Some of these agents have been incorporated into treatment algorithms, while others are currently under investigation demonstrating promising results. This review focuses on highlighting the underlying tumor biology forming the basis of therapeutics in HL, and reviews some of the emerging and established novel therapies.


Subject(s)
Hodgkin Disease , Immunoconjugates , Humans , Hodgkin Disease/pathology , Brentuximab Vedotin/therapeutic use , Immunoconjugates/therapeutic use , Molecular Targeted Therapy/methods , Biology
17.
Pediatr Blood Cancer ; 70(6): e30324, 2023 06.
Article in English | MEDLINE | ID: mdl-37017066

ABSTRACT

BACKGROUND: Although tumor genomic profiling has aided the advancement of targeted genetic therapy, its clinical integration remains a challenge in pediatric cancers due to lower mutation frequency and less available targeted drugs. There have been multiple novel studies examining molecular sequencing in pediatrics; however, many of these studies primarily utilized large-scale, genome-wide screening applications that limit applicable use due to the availability of testing. This study examined the institutional use of a targeted, clinically available approach to tumor genomic sequencing. METHODS: A retrospective chart review was performed on pediatric patients with solid tumors who were managed at Roswell Park Comprehensive Cancer Center and underwent molecular testing of their tumor biopsy via OmniSeq from August 2016 to July 2021. Results were reviewed for mutations considered to be "actionable" by targeted therapy. Patients with actionable mutations were further examined to evaluate treatment course, receival of targeted therapy, and clinical outcomes. RESULTS: We identified 64 pediatric patients consisting of 20 (31%) with CNS tumors and 44 (69%) with non-CNS tumors, ranging in age from 9 months to 21 years. Thirty-five total actionable mutations were identified amongst 27 patients (42%). Of these 27, 12 patients (44%) received at least 1 targeted drug against a respective actionable mutation, of which 6 patients (50%) achieved clinical benefit to therapy, including 1 complete response. CONCLUSIONS: The use of a clinically focused and readily available targeted molecular sequencing panel identified actionable mutations at a comparable rate to the large-scale, less readily available sequencing panels utilized in other studies. Half of our patients who received targeted therapy achieved a complete response or clinical benefit from therapy. Although targeted therapy has a role in pediatric cancer treatment, many newer drugs require further research on their safety and efficacy.


Subject(s)
Neoplasms , Precision Medicine , Humans , Child , Retrospective Studies , Precision Medicine/methods , Neoplasms/drug therapy , Mutation , High-Throughput Nucleotide Sequencing/methods , Genomics/methods , Biomarkers, Tumor/genetics , Molecular Targeted Therapy/methods
18.
Pharmacol Ther ; 245: 108417, 2023 05.
Article in English | MEDLINE | ID: mdl-37075933

ABSTRACT

With the booming development of precision medicine, molecular targeted therapy has been widely used in clinical oncology treatment due to a smaller number of side effects and its superior accuracy compared to that of traditional strategies. Among them, human epidermal growth factor receptor 2 (HER2)-targeted therapy has attracted considerable attention and has been used in the clinical treatment of breast and gastric cancer. Despite excellent clinical effects, HER2-targeted therapy remains in its infancy due to its resulting inherent and acquired resistance. Here, a comprehensive overview of HER2 in numerous cancers is presented, including its biological role, involved signaling pathways, and the status of HER2-targeted therapy.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Female , Humans , Breast Neoplasms/drug therapy , Molecular Targeted Therapy/methods , Signal Transduction
19.
Chin Clin Oncol ; 12(2): 14, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36946186

ABSTRACT

BACKGROUND AND OBJECTIVE: Biliary tract cancers (BTCs), including cholangiocarcinoma and gallbladder cancer, are a relatively rare group of cancers with a poor prognosis. Over the past decade, the utilization of next-generation sequencing has led to the identification of multiple actionable somatic aberrations in BTCs. Subsequently, new therapies have been created to target these molecular alterations and have been incorporated into clinical practice. In this review, we outline therapies that have been previously studied, and those that are under investigation, to target genomic alterations with the goal of improving survival in patients with advanced disease. METHODS: A literature search was performed to identify phase I, II, and III trials of targeted therapies in patients with advanced BTCs published between January 1, 2010 and October 1, 2022. Medline (via PubMed) and ClinicalTrials.gov were searched for relevant studies and 415 trials were identified. The search strategy was performed using keywords including: biliary tract cancer, cholangiocarcinoma, gallbladder cancer, chemotherapy, targeted therapy, randomized trials, controlled trials, phase I, phase II, and phase III. Search results were imported into EndNote X 9.1. KEY CONTENT AND FINDINGS: Overall, immune checkpoint inhibitors, fibroblast growth factor receptor (FGFR) inhibitors, isocitrate dehydrogenase (IDH) inhibitors, and human epidermal growth factor receptor 2 (HER2)-directed therapies have all shown promising results with regard to efficacy in patients with advanced BTCs studied in clinical trials. A number of other agents have also been studied in early-phase trials. CONCLUSIONS: Targeted agents can improve survival in patients with advanced BTCs and have substantially increased the number of potential therapeutic options in patients with refractory disease. The therapeutic landscape of targeted therapies for patients with advanced BTCs continues to evolve based on improvements in detection of genomic alterations.


Subject(s)
Bile Duct Neoplasms , Biliary Tract Neoplasms , Cholangiocarcinoma , Gallbladder Neoplasms , Humans , Gallbladder Neoplasms/drug therapy , Biliary Tract Neoplasms/drug therapy , Biliary Tract Neoplasms/genetics , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Bile Ducts, Intrahepatic , Molecular Targeted Therapy/methods
20.
J Natl Med Assoc ; 115(2): 164-174, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36801148

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is currently the third leading cause of cancer mortality and the incidence is projected to increase by 2030. Despite recent advances in its treatment, African Americans have a 50-60% higher incidence and 30% higher mortality rate when compared to European Americans possibly resulting from differences in socioeconomic status, access to healthcare, and genetics. Genetics plays a role in cancer predisposition, response to cancer therapeutics (pharmacogenetics), and in tumor behavior, making some genes targets for oncologic therapeutics. We hypothesize that the germline genetic differences in predisposition, drug response, and targeted therapies also impact PDAC disparities. To demonstrate the impact of genetics and pharmacogenetics on PDAC disparities, a review of the literature was performed using PubMed with variations of the following keywords: pharmacogenetics, pancreatic cancer, race, ethnicity, African, Black, toxicity, and the FDA-approved drug names: Fluoropyrimidines, Topoisomerase inhibitors, Gemcitabine, Nab-Paclitaxel, Platinum agents, Pembrolizumab, PARP-inhibitors, and NTRK fusion inhibitors. Our findings suggest that the genetic profiles of African Americans may contribute to disparities related to FDA approved chemotherapeutic response for patients with PDAC. We recommend a strong focus on improving genetic testing and participation in biobank sample donations for African Americans. In this way, we can improve our current understanding of genes that influence drug response for patients with PDAC.


Subject(s)
Antineoplastic Agents , Black or African American , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Black or African American/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/ethnology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/ethnology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pharmacogenetics , Molecular Targeted Therapy/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...