Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 95.912
1.
Mol Cancer ; 23(1): 88, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702734

Drug resistance represents a significant obstacle in cancer treatment, underscoring the need for the discovery of novel therapeutic targets. Ubiquitin-specific proteases (USPs), a subclass of deubiquitinating enzymes, play a pivotal role in protein deubiquitination. As scientific research advances, USPs have been recognized as key regulators of drug resistance across a spectrum of treatment modalities, including chemotherapy, targeted therapy, immunotherapy, and radiotherapy. This comprehensive review examines the complex relationship between USPs and drug resistance mechanisms, focusing on specific treatment strategies and highlighting the influence of USPs on DNA damage repair, apoptosis, characteristics of cancer stem cells, immune evasion, and other crucial biological functions. Additionally, the review highlights the potential clinical significance of USP inhibitors as a means to counter drug resistance in cancer treatment. By inhibiting particular USP, cancer cells can become more susceptible to a variety of anti-cancer drugs. The integration of USP inhibitors with current anti-cancer therapies offers a promising strategy to circumvent drug resistance. Therefore, this review emphasizes the importance of USPs as viable therapeutic targets and offers insight into fruitful directions for future research and drug development. Targeting USPs presents an effective method to combat drug resistance across various cancer types, leading to enhanced treatment strategies and better patient outcomes.


Antineoplastic Agents , Drug Resistance, Neoplasm , Neoplasms , Ubiquitin-Specific Proteases , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals , Molecular Targeted Therapy , DNA Repair , Apoptosis/drug effects
2.
Clin Interv Aging ; 19: 681-693, 2024.
Article En | MEDLINE | ID: mdl-38706635

Aging and aging-related diseases present a global public health problem. Therefore, the development of efficient anti-aging drugs has become an important area of research. Traditional Chinese medicine is an important complementary and alternative branch of aging-related diseases therapy. Recently, a growing number of studies have revealed that traditional Chinese medicine has a certain delaying effect on the progression of aging and aging-related diseases. Here, we review the progress in research into using traditional Chinese medicine for aging and aging-related diseases (including neurodegenerative diseases, cardiovascular diseases, diabetes, and cancer). Furthermore, we summarize the potential mechanisms of action of traditional Chinese medicine and provide references for further studies on aging and aging-related diseases.


Aging , Drugs, Chinese Herbal , Medicine, Chinese Traditional , Neoplasms , Neurodegenerative Diseases , Humans , Aging/drug effects , Medicine, Chinese Traditional/methods , Neurodegenerative Diseases/drug therapy , Neoplasms/drug therapy , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Diabetes Mellitus/drug therapy
3.
Int J Nanomedicine ; 19: 3919-3942, 2024.
Article En | MEDLINE | ID: mdl-38708176

Typical physiological characteristics of tumors, such as weak acidity, low oxygen content, and upregulation of certain enzymes in the tumor microenvironment (TME), provide survival advantages when exposed to targeted attacks by drugs and responsive nanomedicines. Consequently, cancer treatment has significantly progressed in recent years. However, the evolution and adaptation of tumor characteristics still pose many challenges for current treatment methods. Therefore, efficient and precise cancer treatments require an understanding of the heterogeneity degree of various factors in cancer cells during tumor evolution to exploit the typical TME characteristics and manage the mutation process. The highly heterogeneous tumor and infiltrating stromal cells, immune cells, and extracellular components collectively form a unique TME, which plays a crucial role in tumor malignancy, including proliferation, invasion, metastasis, and immune escape. Therefore, the development of new treatment methods that can adapt to the evolutionary characteristics of tumors has become an intense focus in current cancer treatment research. This paper explores the latest understanding of cancer evolution, focusing on how tumors use new antigens to shape their "new faces"; how immune system cells, such as cytotoxic T cells, regulatory T cells, macrophages, and natural killer cells, help tumors become "invisible", that is, immune escape; whether the diverse cancer-associated fibroblasts provide support and coordination for tumors; and whether it is possible to attack tumors in reverse. This paper discusses the limitations of targeted therapy driven by tumor evolution factors and explores future strategies and the potential of intelligent nanomedicines, including the systematic coordination of tumor evolution factors and adaptive methods, to meet this therapeutic challenge.


Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/drug effects , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/therapy , Neoplasms/immunology , Nanomedicine/methods , Animals , Nanoparticles/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
4.
Int J Nanomedicine ; 19: 3805-3825, 2024.
Article En | MEDLINE | ID: mdl-38708177

Telomere is a protective structure located at the end of chromosomes of eukaryotes, involved in maintaining the integrity and stability of the genome. Telomeres play an essential role in cancer progression; accordingly, targeting telomere dynamics emerges as an effective approach for the development of cancer therapeutics. Targeting telomere dynamics may work through multifaceted molecular mechanisms; those include the activation of anti-telomerase immune responses, shortening of telomere lengths, induction of telomere dysfunction and constitution of telomerase-responsive drug release systems. In this review, we summarize a wide variety of telomere dynamics-targeted agents in preclinical studies and clinical trials, and reveal their promising therapeutic potential in cancer therapy. As shown, telomere dynamics-active agents are effective as anti-cancer chemotherapeutics and immunotherapeutics. Notably, these agents may display efficacy against cancer stem cells, reducing cancer stem levels. Furthermore, these agents can be integrated with the capability of tumor-specific drug delivery by the constitution of related nanoparticles, antibody drug conjugates and HSA-based drugs.


Antineoplastic Agents , Neoplasms , Telomerase , Telomere , Humans , Neoplasms/drug therapy , Neoplasms/therapy , Telomere/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Telomerase/antagonists & inhibitors , Animals , Drug Delivery Systems/methods , Nanoparticles/chemistry , Immunotherapy/methods , Neoplastic Stem Cells/drug effects
5.
Cell Host Microbe ; 32(5): 627-630, 2024 May 08.
Article En | MEDLINE | ID: mdl-38723599

Microbial-based therapies have the potential to combat immunotherapy resistance, extending the boundaries of oncological therapeutics. In a recent issue of Cell, Jia et al. demonstrates an example of microbial collaboration to produce a postbiotic that promotes the stemness program of CD8+ T cells to augment immunotherapy at the pan-cancer level.


CD8-Positive T-Lymphocytes , Immunotherapy , Neoplasms , Humans , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/drug therapy , CD8-Positive T-Lymphocytes/immunology , Indoles , Animals , Mice
6.
Med Oncol ; 41(6): 145, 2024 May 10.
Article En | MEDLINE | ID: mdl-38727885

Polyelectrolytes represent a unique class of polymers abundant in ionizable functional groups. In a solution, ionized polyelectrolytes can intricately bond with oppositely charged counterparts, giving rise to a fascinating phenomenon known as a polyelectrolyte complex. These complexes arise from the interaction between oppositely charged entities, such as polymers, drugs, and combinations thereof. The polyelectrolyte complexes are highly appealing in cancer management, play an indispensable role in chemotherapy, crafting biodegradable, biocompatible 3D membranes, microcapsules, and nano-sized formulations. These versatile complexes are pivotal in designing controlled and targeted release drug delivery systems. The present review emphasizes on classification of polyelectrolyte complex along with their formation mechanisms. This review comprehensively explores the applications of polyelectrolyte complex, highlighting their efficacy in targeted drug delivery strategies for combating different forms of cancer. The innovative use of polyelectrolyte complex presents a potential breakthrough in cancer therapeutics, demonstrating their role in enhancing treatment precision and effectiveness.


Antineoplastic Agents , Drug Delivery Systems , Neoplasms , Polyelectrolytes , Humans , Polyelectrolytes/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Drug Delivery Systems/methods , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Precision Medicine/methods
7.
PLoS One ; 19(5): e0300425, 2024.
Article En | MEDLINE | ID: mdl-38709807

INTRODUCTION: Deep Venous Thrombosis (DVT) due to Peripherally Inserted Central Catheter (PICC) is one of the most threatening complications after device insertion. OBJECTIVE: To assess the rate of PICC-associated DVT and analyze the risk factors associated with this event in cancer and critically ill patients. METHODS: We conducted a descriptive, retrospective cohort study with 11,588 PICCs from December 2014 to December 2019. Patients ≥ 18 years receiving a PICC were included. Pre-and post-puncture variables were collected and a logistic regression was used to identify the independent factors associated with the risk of DVT. RESULTS: The DVT prevalence was 1.8% (n = 213). The median length of PICC use was 15.3 days. The median age was 75 years (18; 107) and 52% were men, 53.5% were critically ill and 29.1% oncological patients. The most common indications for PICC's were intravenous antibiotics (79.1%). Notably, 91.5% of PICC showed a catheter-to-vein ratio of no more than 33%. The tip location method with intracavitary electrocardiogram was used in 43%. Most catheters (67.9%) were electively removed at the end of intravenous therapy. After adjusting for cancer profile ou chemotherapy, regression anaysis revealed that age (OR 1.011; 95% CI 1.002-1.020), previous DVT (OR 1.96; 95% CI 1.12-3.44) and obstruction of the device (OR 1.60; 95% CI 1.05-2.42) were independent factors associated with PICC-associated DVT, whereas the use of an anticoagulant regimen was a protective variable (OR 0.73; 95% CI 0.54-0.99). CONCLUSION: PICC is a safe and suitable intravenous device for medium and long-term therapy, with low rates of DVT even in a cohort of critically ill and cancer patients.


Catheterization, Peripheral , Venous Thrombosis , Humans , Male , Female , Aged , Retrospective Studies , Venous Thrombosis/etiology , Venous Thrombosis/epidemiology , Middle Aged , Brazil/epidemiology , Catheterization, Peripheral/adverse effects , Risk Factors , Adult , Aged, 80 and over , Neoplasms/complications , Neoplasms/drug therapy , Catheterization, Central Venous/adverse effects , Critical Illness , Adolescent , Young Adult , Prevalence
8.
JCO Clin Cancer Inform ; 8: e2400051, 2024 May.
Article En | MEDLINE | ID: mdl-38713889

This new editorial discusses the promise and challenges of successful integration of natural language processing methods into electronic health records for timely, robust, and fair oncology pharmacovigilance.


Artificial Intelligence , Electronic Health Records , Medical Oncology , Natural Language Processing , Pharmacovigilance , Humans , Medical Oncology/methods , Data Collection/methods , Neoplasms/drug therapy , Adverse Drug Reaction Reporting Systems
9.
Acta Oncol ; 63: 313-321, 2024 May 08.
Article En | MEDLINE | ID: mdl-38716486

BACKGROUND: A significant proportion of patients with incurable cancer receive systemic anticancer therapy (SACT) within their last 30 days of life (DOL). The treatment has questionable benefit, nevertheless is considered a quality indicator of end-of-life (EOL) care. This retrospective cohort study aims to investigate the rates and potential predictors of SACT and factors associated with SACT within the last 30 DOL. The study also evaluates the scope of Eastern Cooperative Oncology Group (ECOG) performance status and the modified Glasgow prognostic score (mGPS) as decision-making tools for oncologists. PATIENTS AND MATERIAL: This review of medical records included 383 patients with non-curable cancer who died between July 2018 and December 2019. Descriptive statistics with Chi-squared tests and regression analysis were used to identify factors associated with SACT within the last 30 DOL. RESULTS: Fifty-seven (15%) patients received SACT within the last 30 DOL. SACT within 30 last DOL was associated with shorter time from diagnosis until death (median 234 days vs. 482, p = 0.008) and ECOG score < 3 30 days prior to death (p = 0.001). Patients receiving SACT during the last 30 DOL were more likely to be hospitalised and die in hospital. ECOG and mGPS score were stated at start last line of treatment only in 139 (51%) and 135 (49%) respectively. INTERPRETATION: Those with short time since diagnosis tended to receive SACT more frequently the last 30 DOL. The use of mGPS as a decision-making tool is modest, and there is lack in documentation of performance status.


Neoplasms , Terminal Care , Humans , Retrospective Studies , Male , Female , Neoplasms/drug therapy , Neoplasms/mortality , Neoplasms/therapy , Aged , Terminal Care/methods , Middle Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Adult , Prognosis , Palliative Care/methods , Palliative Care/statistics & numerical data
11.
Nat Commun ; 15(1): 3860, 2024 May 08.
Article En | MEDLINE | ID: mdl-38719824

Dual blocker therapy (DBT) has the enhanced antitumor benefits than the monotherapy. Yet, few effective biomarkers are developed to monitor the therapy response. Herein, we investigate the DBT longitudinal plasma proteome profiling including 113 longitudinal samples from 22 patients who received anti-PD1 and anti-CTLA4 DBT therapy. The results show the immune response and cholesterol metabolism are upregulated after the first DBT cycle. Notably, the cholesterol metabolism is activated in the disease non-progressive group (DNP) during the therapy. Correspondingly, the clinical indicator prealbumin (PA), free triiodothyronine (FT3) and triiodothyronine (T3) show significantly positive association with the cholesterol metabolism. Furthermore, by integrating proteome and radiology approach, we observe the high-density lipoprotein partial remodeling are activated in DNP group and identify a candidate biomarker APOC3 that can reflect DBT response. Above, we establish a machine learning model to predict the DBT response and the model performance is validated by an independent cohort with balanced accuracy is 0.96. Thus, the plasma proteome profiling strategy evaluates the alteration of cholesterol metabolism and identifies a panel of biomarkers in DBT.


Cholesterol , Proteome , Humans , Cholesterol/blood , Cholesterol/metabolism , Proteome/metabolism , Female , Male , Middle Aged , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/metabolism , CTLA-4 Antigen/blood , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/blood , Biomarkers/blood , Aged , Triiodothyronine/blood , Machine Learning , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/blood , Neoplasms/metabolism , Proteomics/methods
12.
Article En | MEDLINE | ID: mdl-38728674

Objective: To examine the complexities of psychotropic medication prescription in home-based palliative care for oncology patients.Methods: A retrospective analysis of 125 medical records of patients receiving palliative home care for cancer was conducted at a tertiary hospital, with a specific focus on the prescription patterns of psychotropic medications. The data were collected in September 2023.Results: Among 125 cases, the mean age was 64.4 ± 14.9 years, with 50.4% females. Breast cancer (14.4%) and lung cancer (13.6%) were the most common diagnoses. Psychotropic medication was administered to 35.2% of patients. Treatment was initiated by palliative care doctors in 75% of cases, while psychiatrists handled 25%. Medication selection was predominantly symptom driven (63%), with anxiety prompting benzodiazepine prescriptions in 50% of cases, depression resulting in antidepressant use in 22%, and psychosis leading to antipsychotic treatment in 18%. Specific diagnoses were the target in only 36% of prescriptions, with delirium (27%) being the most prevalent, followed by depression and bipolar disorder. Benzodiazepines were the most commonly prescribed class of medications (56.8%), with clonazepam being the most prevalent (40.9%), followed by alprazolam and lorazepam (15.9%). Atypical antipsychotics made up 43.1% of prescriptions, with quetiapine being the most frequently prescribed (34%), along with olanzapine and risperidone (11%). Antidepressants accounted for 31.8% of prescriptions, including selective serotonin reuptake inhibitors at 18% and mirtazapine and amitriptyline at 6% each. Haloperidol, a typical antipsychotic, was prescribed in 13.6% of cases. Polypharmacy was observed in 35.6% of patients.Conclusion: In palliative home care, psychotropic medications are frequently prescribed by palliative doctors primarily for symptom management, with limited psychiatric consultations and challenges in accessing psychological evaluations. Collaborative efforts among regional or institutional medical bodies, including psychiatrists, psychologists, palliative doctors, and social workers, are needed to establish ethical guidelines for appropriate and effective psychotropic prescription.Prim Care Companion CNS Disord 2024;26(2):23m03668. Author affiliations are listed at the end of this article.


Home Care Services , Neoplasms , Palliative Care , Psychotropic Drugs , Humans , Female , Male , Middle Aged , Retrospective Studies , Psychotropic Drugs/therapeutic use , Aged , Neoplasms/drug therapy , Neoplasms/complications , Drug Prescriptions/statistics & numerical data , Adult , Aged, 80 and over , Practice Patterns, Physicians'/statistics & numerical data
13.
Br Dent J ; 236(9): 683-687, 2024 May.
Article En | MEDLINE | ID: mdl-38730156

The continuation of bone-modifying agents (BMAs) in patients with established medication-related osteonecrosis of the jaw (MRONJ) is a common concern among dentists and oncologists. There is little evidence supporting or refuting the continued use of BMAs or drug holidays and their impact on established MRONJ. This paper evaluates the outcome of continued BMAs use on the patient's MRONJ status. A retrospective review of 29 oncology patients undergoing active cancer care for either metastatic disease or multiple myeloma was conducted. Data on demographics, oncological status, BMA history and MRONJ status were collected. In total, 90% of patients were judged to have healed or stable MRONJ while continuing BMAs. Most patients (69%) continued the same BMA regime (three- or four-weekly) that they were on before developing MRONJ. The average number of BMAs doses received after an MRONJ diagnosis was 12 (range 1-48). Three patients (10.3%) were found to have MRONJ progression, with two patients developing new sites of necrosis. This real-world dataset suggests that the majority of MRONJ cases remain stable and will not worsen with the continuation of BMAs.


Bisphosphonate-Associated Osteonecrosis of the Jaw , Bone Density Conservation Agents , Humans , Bisphosphonate-Associated Osteonecrosis of the Jaw/etiology , Retrospective Studies , Male , Female , Middle Aged , Bone Density Conservation Agents/adverse effects , Bone Density Conservation Agents/therapeutic use , Aged , Aged, 80 and over , Adult , Multiple Myeloma/drug therapy , Neoplasms/drug therapy
14.
Biomed Mater ; 19(4)2024 May 10.
Article En | MEDLINE | ID: mdl-38697132

During the process of malignant tumor treatment, photodynamic therapy (PDT) exerts poor efficacy due to the hypoxic environment of the tumor cells, and long-time chemotherapy reduces the sensitivity of tumor cells to chemotherapy drugs due to the presence of drug-resistant proteins on the cell membranes for drug outward transportation. Therefore, we reported a nano platform based on mesoporous silica coated with polydopamine (MSN@PDA) loading PDT enhancer MnO2, photosensitizer indocyanine green (ICG) and chemotherapeutic drug doxorubicin (DOX) (designated as DMPIM) to achieve a sequential release of different drugs to enhance treatment of malignant tumors. MSN was first synthesized by a template method, then DOX was loaded into the mesoporous channels of MSN, and locked by the PDA coating. Next, ICG was modified by π-π stacking on PDA, and finally, MnO2layer was accumulated on the surface of DOX@MSN@PDA- ICG@MnO2, achieving orthogonal loading and sequential release of different drugs. DMPIM first generated oxygen (O2) through the reaction between MnO2and H2O2after entering tumor cells, alleviating the hypoxic environment of tumors and enhancing the PDT effect of sequentially released ICG. Afterwards, ICG reacted with O2in tumor tissue to produce reactive oxygen species, promoting lysosomal escape of drugs and inactivation of p-glycoprotein (p-gp) on tumor cell membranes. DOX loaded in the MSN channels exhibited a delay of approximately 8 h after ICG release to exert the enhanced chemotherapy effect. The drug delivery system achieved effective sequential release and multimodal combination therapy, which achieved ideal therapeutic effects on malignant tumors. This work offers a route to a sequential drug release for advancing the treatment of malignant tumors.


Doxorubicin , Drug Liberation , Indocyanine Green , Indoles , Manganese Compounds , Oxides , Photochemotherapy , Photosensitizing Agents , Polymers , Photochemotherapy/methods , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Indocyanine Green/chemistry , Indoles/chemistry , Animals , Manganese Compounds/chemistry , Humans , Polymers/chemistry , Cell Line, Tumor , Oxides/chemistry , Photosensitizing Agents/chemistry , Silicon Dioxide/chemistry , Mice , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Drug Delivery Systems , Nanoparticles/chemistry , Drug Carriers/chemistry , Porosity
15.
J Inorg Biochem ; 256: 112569, 2024 Jul.
Article En | MEDLINE | ID: mdl-38701687

The clinical success of [223Ra]RaCl2 (Xofigo®) for the palliative treatment of bone metastases in patients with prostate cancer has highlighted the therapeutic potential of α-particle emission. Expanding the applicability of radium-223 in Targeted Alpha Therapy of non-osseous tumors is followed up with significant interest, as it holds the potential to unveil novel treatment options in the comprehensive management of cancer. Moreover, the use of barium radionuclides, like barium-131 and -135m, is still unfamiliar in nuclear medicine applications, although they can be considered as radium-223 surrogates for imaging purposes. Enabling these applications requires the establishment of chelators able to form stable complexes with radium and barium radionuclides. Until now, only a limited number of ligands have been suggested and these molecules have been primarily inspired by existing structures known for their ability to complex large metal cations. However, a systematic inspection of chelators specifically tailored to Ra2+ and Ba2+ has yet to be conducted. This work delves into a comprehensive investigation of a series of small organic ligands, aiming to unveil the coordination preferences of both radium-223 and barium-131/135m. Electronic binding energies of both metal cations to each ligand were theoretically computed via Density Functional Theory calculations (COSMO-ZORA-PBE-D3/TZ2P), while thermodynamic stability constants were experimentally determined for Ba2+-ligand complexes by potentiometry, NMR and UV-Vis spectroscopies. The outcomes revealed malonate, 2-hydroxypyridine 1-oxide and picolinate as the most favorable building blocks to design multidentate chelators. These findings serve as foundation guidelines, propelling the development of cutting-edge radium-223- and barium-131/135m-based radiopharmaceuticals for Targeted Alpha Therapy and theranostics of cancer.


Radium , Radium/chemistry , Radium/therapeutic use , Humans , Radioisotopes/chemistry , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use , Barium/chemistry , Alpha Particles/therapeutic use , Chelating Agents/chemistry , Chelating Agents/therapeutic use , Neoplasms/drug therapy , Theranostic Nanomedicine/methods , Metals, Alkaline Earth/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/therapeutic use
16.
J Cancer Res Clin Oncol ; 150(5): 237, 2024 May 07.
Article En | MEDLINE | ID: mdl-38713378

PURPOSE: Bispecific antibodies (BsAbs), capable of targeting two antigens simultaneously, represent a significant advancement by employing dual mechanisms of action for tumor suppression. However, how to pair targets to develop effective and safe bispecific drugs is a major challenge for pharmaceutical companies. METHODS: Using machine learning models, we refined the biological characteristics of currently approved or in clinical development BsAbs and analyzed hundreds of membrane proteins as bispecific targets to predict the likelihood of successful drug development for various target combinations. Moreover, to enhance the interpretability of prediction results in bispecific target combination, we combined machine learning models with Large Language Models (LLMs). Through a Retrieval-Augmented Generation (RAG) approach, we supplement each pair of bispecific targets' machine learning prediction with important features and rationales, generating interpretable analytical reports. RESULTS: In this study, the XGBoost model with pairwise learning was employed to predict the druggability of BsAbs. By analyzing extensive data on BsAbs and designing features from perspectives such as target activity, safety, cell type specificity, pathway mechanism, and gene embedding representation, our model is able to predict target combinations of BsAbs with high market potential. Specifically, we integrated XGBoost with the GPT model to discuss the efficacy of each bispecific target pair, thereby aiding the decision-making for drug developers. CONCLUSION: The novelty of this study lies in the integration of machine learning and GPT techniques to provide a novel framework for the design of BsAbs drugs. This holistic approach not only improves prediction accuracy, but also enhances the interpretability and innovativeness of drug design.


Antibodies, Bispecific , Machine Learning , Antibodies, Bispecific/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/immunology
17.
J Nanobiotechnology ; 22(1): 228, 2024 May 08.
Article En | MEDLINE | ID: mdl-38715049

Development of ferroptosis-inducible nanoplatforms with high efficiency and specificity is highly needed and challenging in tumor ferrotherapy. Here, we demonstrate highly effective tumor ferrotherapy using iron (II)-based metal-organic framework (FessMOF) nanoparticles, assembled from disulfide bonds and ferrous ions. The as-prepared FessMOF nanoparticles exhibit peroxidase-like activity and pH/glutathione-dependent degradability, which enables tumor-responsive catalytic therapy and glutathione depletion by the thiol/disulfide exchange to suppress glutathione peroxidase 4, respectively. Upon PEGylation and Actinomycin D (ActD) loading, the resulting FessMOF/ActD-PEG nanoplatform induces marked DNA damage and lipid peroxidation. Concurrently, we found that ActD can inhibit Xc- system and elicit ferritinophagy, which further boosts the ferrotherapeutic efficacy of the FessMOF/ActD-PEG. In vivo experiments demonstrate that our fabricated nanoplatform presents excellent biocompatibility and a high tumor inhibition rate of 91.89%.


DNA Damage , Ferroptosis , Iron , Metal-Organic Frameworks , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Ferroptosis/drug effects , Animals , Humans , Mice , DNA Damage/drug effects , Iron/chemistry , Cell Line, Tumor , DNA Repair/drug effects , Nanoparticles/chemistry , Neoplasms/drug therapy , Mice, Inbred BALB C , Female
18.
J Exp Med ; 221(7)2024 Jul 01.
Article En | MEDLINE | ID: mdl-38709209

New studies (Tang et al. 2024. J. Exp. Med.https://doi.org/10.1084/jem.20231395) describe a liver stress pathway that is activated by certain chemotherapeutic drugs, which in turn induces a peptide hormone which partially mediates the lower food intake and body weight loss during chemotherapy treatment.


Antineoplastic Agents , Humans , Antineoplastic Agents/therapeutic use , Liver/metabolism , Animals , Stress, Physiological , Neoplasms/drug therapy
19.
Oncotarget ; 15: 275-284, 2024 May 03.
Article En | MEDLINE | ID: mdl-38709242

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and COVID-19 infection has led to worsened outcomes for patients with cancer. SARS-CoV-2 spike protein mediates host cell infection and cell-cell fusion that causes stabilization of tumor suppressor p53 protein. In-silico analysis previously suggested that SARS-CoV-2 spike interacts with p53 directly but this putative interaction has not been demonstrated in cells. We examined the interaction between SARS-CoV-2 spike, p53 and MDM2 (E3 ligase, which mediates p53 degradation) in cancer cells using an immunoprecipitation assay. We observed that SARS-CoV-2 spike protein interrupts p53-MDM2 protein interaction but did not detect SARS-CoV-2 spike bound with p53 protein in the cancer cells. We further observed that SARS-CoV-2 spike suppresses p53 transcriptional activity in cancer cells including after nutlin exposure of wild-type p53-, spike-expressing tumor cells and inhibits chemotherapy-induced p53 gene activation of p21(WAF1), TRAIL Death Receptor DR5 and MDM2. The suppressive effect of SARS-CoV-2 spike on p53-dependent gene activation provides a potential molecular mechanism by which SARS-CoV-2 infection may impact tumorigenesis, tumor progression and chemotherapy sensitivity. In fact, cisplatin-treated tumor cells expressing spike were found to have increased cell viability as compared to control cells. Further observations on γ-H2AX expression in spike-expressing cells treated with cisplatin may indicate altered DNA damage sensing in the DNA damage response pathway. The preliminary observations reported here warrant further studies to unravel the impact of SARS-CoV-2 and its various encoded proteins including spike on pathways of tumorigenesis and response to cancer therapeutics. More efforts should be directed at studying the effects of the SARS-CoV-2 spike and other viral proteins on host DNA damage sensing, response and repair mechanisms. A goal would be to understand the structural basis for maximal anti-viral immunity while minimizing suppression of host defenses including the p53 DNA damage response and tumor suppression pathway. Such directions are relevant and important including not only in the context of viral infection and mRNA vaccines in general but also for patients with cancer who may be receiving cytotoxic or other cancer treatments.


Cell Survival , Cyclin-Dependent Kinase Inhibitor p21 , Proto-Oncogene Proteins c-mdm2 , Receptors, TNF-Related Apoptosis-Inducing Ligand , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Tumor Suppressor Protein p53 , Humans , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Cell Survival/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , SARS-CoV-2/physiology , Cell Line, Tumor , Neoplasms/metabolism , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Transfection , COVID-19/virology , COVID-19/metabolism
...