1
|
Patlolla SS, Alam AH, Katz JN, Hall SA. Tailored Therapy in Cardiogenic Shock: Case-Based Management Choices. Am J Cardiol 2025; 235:30-36. [PMID: 39489269 DOI: 10.1016/j.amjcard.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
Cardiogenic shock (CS) is a complex, multisystem disorder precipitated by hypoperfusion from cardiac dysfunction. Our current approach to defining and treating CS encompasses all patients under 1 umbrella regardless of phenotype. This has created challenges for clinical trials and patient care owing to the heterogeneity of the patient population with CS. The Society of Coronary Angiography and Interventions shock classification has created a universal language for CS that has been rapidly adopted by researchers and clinicians. Its latest iteration established the 3-axis model incorporating shock severity, risk modifiers, and phenotypes. Phenotypes of CS have unique hemodynamic profiles that require nuanced adjustment approaches. In this study, we discuss representative cases including acute myocardial infarction, acute-on-chronic heart failure, fulminant myocarditis, and right ventricular failure. For each phenotype, specific hemodynamic parameters may help confirm appropriate diagnosis and direct to therapeutic targets signaling stability and recovery. The underlying pathophysiology of each phenotype can also help predict the extent of stabilization with pharmacologic interventions or the need to escalate to mechanical circulatory support. In conclusion, this tailored approach to CS, rather than a 1-size-fits-all approach, could help improve outcomes.
Collapse
Affiliation(s)
- S Shiva Patlolla
- Center for Advanced Heart and Lung Disease, Baylor University Medical Center, Dallas, Texas.
| | - Amit H Alam
- Division of Cardiology, New York University Langone Health, New York, New York
| | - Jason N Katz
- Division of Cardiology, New York University Langone Health, New York, New York
| | - Shelley A Hall
- Center for Advanced Heart and Lung Disease, Baylor University Medical Center, Dallas, Texas
| |
Collapse
|
2
|
Khilwani R, Singh S. Leveraging Evolutionary Immunology in Interleukin-6 and Interleukin-17 Signaling for Lung Cancer Therapeutics. ACS Pharmacol Transl Sci 2024; 7:3658-3670. [PMID: 39698267 PMCID: PMC11650734 DOI: 10.1021/acsptsci.4c00412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
Lung cancer is among the most common instances of cancer subtypes and is associated with high mortality rates. Due to the availability of fewer therapies and delayed clinical investigations, the number of cancer incidences is rising dramatically. This is possibly an effect of immune modulations and chemotherapeutic drugs that raises cancer resistance. Among the list, IL-6 and IL-17 are host-derived paradoxical effectors that attune immune responses in malignant lung cells. Their excessive release in the cytokine milieu stabilizes immunosuppressive phenotypes, resulting in cellular perturbations. During tumor development, the significance of these molecules is reflected in their potential to regulate oncogenesis by initiating a myriad of signaling events that influence tumor growth and the metastatic ability of benign cancer cells. Moreover, their transactivation contributes to antiapoptotic mechanisms and favors cancer cell survival via constitutive expression of immunoregulatory molecules. Co-evolution and gene duplication events could be the major drivers behind cytokine evolution, which have prompted generic changes and, hence, the additive effect. The evolutionary model and statistical analysis provide evidence about the cytokines ancestral relationships and site-specific conservation, which is more convincing as both cytokines share cysteine-knot-like structures important in maintaining structural integrity. Funneling through the findings could help find residues that serve a catalytic role in immune functioning. Designing peptides or subunit vaccine formulations against those conserved residues could aid in combating lung cancer pathogenesis.
Collapse
Affiliation(s)
- Riya Khilwani
- Systems Medicine Laboratory, BRIC-National Centre for Cell Science, NCCS Complex,
Ganeshkhind, SPPU Campus, Pune 411007, India
| | - Shailza Singh
- Systems Medicine Laboratory, BRIC-National Centre for Cell Science, NCCS Complex,
Ganeshkhind, SPPU Campus, Pune 411007, India
| |
Collapse
|
3
|
Fionda B, Placidi E, de Ridder M, Strigari L, Patarnello S, Tanderup K, Hannoun-Levi JM, Siebert FA, Boldrini L, Antonietta Gambacorta M, De Spirito M, Sala E, Tagliaferri L. Artificial intelligence in interventional radiotherapy (brachytherapy): Enhancing patient-centered care and addressing patients' needs. Clin Transl Radiat Oncol 2024; 49:100865. [PMID: 39381628 PMCID: PMC11459626 DOI: 10.1016/j.ctro.2024.100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
This review explores the integration of artificial intelligence (AI) in interventional radiotherapy (IRT), emphasizing its potential to streamline workflows and enhance patient care. Through a systematic analysis of 78 relevant papers spanning from 2002 to 2024, we identified significant advancements in contouring, treatment planning, outcome prediction, and quality assurance. AI-driven approaches offer promise in reducing procedural times, personalizing treatments, and improving treatment outcomes for oncological patients. However, challenges such as clinical validation and quality assurance protocols persist. Nonetheless, AI presents a transformative opportunity to optimize IRT and meet evolving patient needs.
Collapse
Affiliation(s)
- Bruno Fionda
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Elisa Placidi
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Mischa de Ridder
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lidia Strigari
- Department of Medical Physics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Stefano Patarnello
- Real World Data Facility, Gemelli Generator, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Kari Tanderup
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jean-Michel Hannoun-Levi
- Department of Radiation Oncology, Antoine Lacassagne Cancer Centre, University of Côte d’Azur, Nice, France
| | - Frank-André Siebert
- Clinic of Radiotherapy (Radiooncology), University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Luca Boldrini
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Maria Antonietta Gambacorta
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco De Spirito
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
- Dipartimento di Neuroscienze, Sezione di Fisica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Evis Sala
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Tagliaferri
- Dipartimento di Diagnostica per Immagini e Radioterapia Oncologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
4
|
Stewart GM, Tong BK, Cistulli PA. Precision medicine approaches in obstructive sleep apnoea: The role of dentist-sleep physician partnerships. Aust Dent J 2024. [PMID: 39354705 DOI: 10.1111/adj.13039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2024] [Indexed: 10/03/2024]
Abstract
Obstructive Sleep Apnoea (OSA) is a common heterogenous sleep disorder that is associated with a wide range of comorbidities and consequences, including the development of neurocognitive and cardiometabolic disorders. The heterogeneity of OSA necessitates a precision medicine approach to accurately diagnose this condition and to effectively manage patients. One of the primary models of precision medicine is described by the P4 approach of predicting those who are susceptible to disease, preventing the occurrence of disease, personalizing treatment, and encouraging patients to participate in their individual healthcare journey. Recent advances in oral appliance therapy and OSA monitoring techniques have fostered an exciting opportunity for enhanced collaboration between dentists and sleep physicians to optimize OSA precision medicine care. This review aims to discuss the sources of heterogeneity among OSA patients, provide an overview of the growing applications of oral appliance therapy and tailored monitoring programs for OSA that are shifting treatment to a more personalized and participatory model of care, and outline the pivotal role of dentists in managing patients with OSA.
Collapse
Affiliation(s)
- G M Stewart
- Charles Perkins Centre and Sydney Medical School, University of Sydney, Camperdown, Sydney, Australia
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - B K Tong
- Charles Perkins Centre and Sydney Medical School, University of Sydney, Camperdown, Sydney, Australia
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| | - P A Cistulli
- Charles Perkins Centre and Sydney Medical School, University of Sydney, Camperdown, Sydney, Australia
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, St Leonards, Sydney, Australia
| |
Collapse
|
5
|
López Rivera JJ, Rueda-Gaitán P, Rios Pinto LC, Rodríguez Gutiérrez DA, Gomez-Lopera N, Lamilla J, Rojas Aguirre FA, Bernal Vaca L, Isaza-Ruget MA. Advancing Cancer Care in Colombia: Results of the First In Situ Implementation of Comprehensive Genomic Profiling. J Pers Med 2024; 14:975. [PMID: 39338229 PMCID: PMC11433056 DOI: 10.3390/jpm14090975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Comprehensive genomic profiling (CGP) identifies genetic alterations and patterns that are crucial for therapy selection and precise treatment development. In Colombia, limited access to CGP tests underscores the necessity of documenting the prevalence of treatable genetic alterations. This study aimed to describe the somatic genetic profile of specific cancer types in Colombian patients and assess its impact on treatment selection. METHODS A retrospective cohort study was conducted at Clínica Colsanitas S.A. from March 2023 to June 2024. Sequencing was performed on the NextSeq2000 platform with the TruSight Oncology 500 (TSO500) assay, which simultaneously evaluates 523 genes for DNA analysis and 55 for RNA; additionally, analyses were performed with the SOPHiA DDM software. The tumor mutational burden (TMB), microsatellite instability (MSI), and programmed cell death ligand 1 (PDL1) were assessed. RESULTS Among 111 patients, 103 were evaluated, with gastrointestinal (27.93%), respiratory (13.51%), and central nervous system cancers (10.81%) being the most prevalent. TP53 (37%), KMT2C (28%), and KRAS (21%) were frequent mutations. Actionable findings were detected in 76.7% of cases, notably in digestive (20 patients) and lung cancers (8 patients). MSI was stable at 82.52% and high at 2.91%, whilst TMB was predominantly low (91.26%). CONCLUSIONS The test has facilitated access to targeted therapies, improving clinical outcomes in Colombian patients. This profiling test is expected to increase opportunities for personalized medicine in Colombia.
Collapse
Affiliation(s)
- Juan Javier López Rivera
- Laboratorio Clínico Especializado, Clínica Universitaria Colombia, Clínica Colsanitas, Bogotá 111321, Colombia
- Grupo de Genética Médica, Clínica Universitaria Colombia, Clínica Colsanitas, Bogotá 111321, Colombia
| | - Paula Rueda-Gaitán
- Laboratorio Clínico Especializado, Clínica Universitaria Colombia, Clínica Colsanitas, Bogotá 111321, Colombia
| | - Laura Camila Rios Pinto
- Laboratorio Clínico Especializado, Clínica Universitaria Colombia, Clínica Colsanitas, Bogotá 111321, Colombia
| | | | - Natalia Gomez-Lopera
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá 111321, Colombia
| | - Julian Lamilla
- Laboratorio Clínico Especializado, Clínica Universitaria Colombia, Clínica Colsanitas, Bogotá 111321, Colombia
| | | | - Laura Bernal Vaca
- Servicio de Oncología, Clínica Universitaria Colombia, Clínica Colsanitas, Bogotá 111321, Colombia
| | - Mario Arturo Isaza-Ruget
- Laboratorio Clínico y de Patología, Clínica Colsanitas, Grupo Keralty, Bogotá 111321, Colombia
- Grupo de Investigación en Patología Clínica (INPAC), Fundación Universitaria Sanitas, Bogotá 111321, Colombia
| |
Collapse
|
6
|
Basar E, Mead H, Shum B, Rauter I, Ay C, Skaletz-Rorowski A, Brockmeyer NH. Biological Barriers for Drug Delivery and Development of Innovative Therapeutic Approaches in HIV, Pancreatic Cancer, and Hemophilia A/B. Pharmaceutics 2024; 16:1207. [PMID: 39339243 PMCID: PMC11435036 DOI: 10.3390/pharmaceutics16091207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024] Open
Abstract
Biological barriers remain a major obstacle for the development of innovative therapeutics. Depending on a disease's pathophysiology, the involved tissues, cell populations, and cellular components, drugs often have to overcome several biological barriers to reach their target cells and become effective in a specific cellular compartment. Human biological barriers are incredibly diverse and include multiple layers of protection and obstruction. Importantly, biological barriers are not only found at the organ/tissue level, but also include cellular structures such as the outer plasma membrane, the endolysosomal machinery, and the nuclear envelope. Nowadays, clinicians have access to a broad arsenal of therapeutics ranging from chemically synthesized small molecules, biologicals including recombinant proteins (such as monoclonal antibodies and hormones), nucleic-acid-based therapeutics, and antibody-drug conjugates (ADCs), to modern viral-vector-mediated gene therapy. In the past decade, the therapeutic landscape has been changing rapidly, giving rise to a multitude of innovative therapy approaches. In 2018, the FDA approval of patisiran paved the way for small interfering RNAs (siRNAs) to become a novel class of nucleic-acid-based therapeutics, which-upon effective drug delivery to their target cells-allow to elegantly regulate the post-transcriptional gene expression. The recent approvals of valoctocogene roxaparvovec and etranacogene dezaparvovec for the treatment of hemophilia A and B, respectively, mark the breakthrough of viral-vector-based gene therapy as a new tool to cure disease. A multitude of highly innovative medicines and drug delivery methods including mRNA-based cancer vaccines and exosome-targeted therapy is on the verge of entering the market and changing the treatment landscape for a broad range of conditions. In this review, we provide insights into three different disease entities, which are clinically, scientifically, and socioeconomically impactful and have given rise to many technological advancements: acquired immunodeficiency syndrome (AIDS) as a predominant infectious disease, pancreatic carcinoma as one of the most lethal solid cancers, and hemophilia A/B as a hereditary genetic disorder. Our primary objective is to highlight the overarching principles of biological barriers that can be identified across different disease areas. Our second goal is to showcase which therapeutic approaches designed to cross disease-specific biological barriers have been promising in effectively treating disease. In this context, we will exemplify how the right selection of the drug category and delivery vehicle, mode of administration, and therapeutic target(s) can help overcome various biological barriers to prevent, treat, and cure disease.
Collapse
Affiliation(s)
- Emre Basar
- WIR—Walk In Ruhr, Center for Sexual Health & Medicine, Department of Dermatology, Venerology and Allergology, Ruhr-University Bochum, 44787 Bochum, Germany;
| | | | - Bennett Shum
- GenePath LLC, Sydney, NSW 2067, Australia
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of NSW, Sydney, NSW 2052, Australia
| | | | - Cihan Ay
- Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria
| | - Adriane Skaletz-Rorowski
- WIR—Walk In Ruhr, Center for Sexual Health & Medicine, Department of Dermatology, Venerology and Allergology, Ruhr-University Bochum, 44787 Bochum, Germany;
| | - Norbert H. Brockmeyer
- WIR—Walk In Ruhr, Center for Sexual Health & Medicine, Department of Dermatology, Venerology and Allergology, Ruhr-University Bochum, 44787 Bochum, Germany;
| |
Collapse
|
7
|
Mao W, Zhou T, Zhang F, Qian M, Xie J, Li Z, Shu Y, Li Y, Xu H. Pan-cancer single-cell landscape of drug-metabolizing enzyme genes. Pharmacogenet Genomics 2024; 34:217-225. [PMID: 38814173 DOI: 10.1097/fpc.0000000000000538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
OBJECTIVE Varied expression of drug-metabolizing enzymes (DME) genes dictates the intensity and duration of drug response in cancer treatment. This study aimed to investigate the transcriptional profile of DMEs in tumor microenvironment (TME) at single-cell level and their impact on individual responses to anticancer therapy. METHODS Over 1.3 million cells from 481 normal/tumor samples across 9 solid cancer types were integrated to profile changes in the expression of DME genes. A ridge regression model based on the PRISM database was constructed to predict the influence of DME gene expression on drug sensitivity. RESULTS Distinct expression patterns of DME genes were revealed at single-cell resolution across different cancer types. Several DME genes were highly enriched in epithelial cells (e.g. GPX2, TST and CYP3A5 ) or different TME components (e.g. CYP4F3 in monocytes). Particularly, GPX2 and TST were differentially expressed in epithelial cells from tumor samples compared to those from normal samples. Utilizing the PRISM database, we found that elevated expression of GPX2, CYP3A5 and reduced expression of TST was linked to enhanced sensitivity of particular chemo-drugs (e.g. gemcitabine, daunorubicin, dasatinib, vincristine, paclitaxel and oxaliplatin). CONCLUSION Our findings underscore the varied expression pattern of DME genes in cancer cells and TME components, highlighting their potential as biomarkers for selecting appropriate chemotherapy agents.
Collapse
Affiliation(s)
- Wei Mao
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan
| | - Tao Zhou
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan
| | - Feng Zhang
- Center for Precision Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and Oncology, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai
| | - Jianqiang Xie
- Department of Medicine and Surgery, Sichan Second Veterans Hospital
| | - Zhengyan Li
- Department of Radiology, West China Hospital, Sichuan University
| | - Yang Shu
- Gastric Cancer Center, West China Hospital, Sichuan University
| | - Yuan Li
- Institute of Digestive Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Heng Xu
- Department of Laboratory Medicine/Research Centre of Clinical Laboratory Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan
| |
Collapse
|
8
|
Lin L, van der Noort V, Steeghs N, Ruiter G, Beijnen JH, Huitema ADR. A joint model of longitudinal pharmacokinetic and time-to-event data to study exposure-response relationships: a proof-of-concept study with alectinib. Cancer Chemother Pharmacol 2024; 94:453-459. [PMID: 38992166 PMCID: PMC11420381 DOI: 10.1007/s00280-024-04698-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
PURPOSE In exposure-response analyses of oral targeted anticancer agents, longitudinal plasma trough concentrations are often aggregated into a single value even though plasma trough concentrations can vary over time due to dose adaptations, for example. The aim of this study was to compare joint models to conventional exposure-response analyses methods with the application of alectinib as proof-of-concept. METHODS Joint models combine longitudinal pharmacokinetic data and progression-free survival data to infer the dependency and association between the two datatypes. The results from the best joint model and the standard and time-dependent cox proportional hazards models were compared. To normalize the data, alectinib trough concentrations were normalized using a sigmoidal transformation to transformed trough concentrations (TTC) before entering the models. RESULTS No statistically significant exposure-response relationship was observed in the different Cox models. In contrast, the joint model with the current value of TTC in combination with the average TTC over time did show an exposure-response relationship for alectinib. A one unit increase in the average TTC corresponded to an 11% reduction in progression (HR, 0.891; 95% confidence interval, 0.805-0.988). CONCLUSION Joint models are able to give insights in the association structure between plasma trough concentrations and survival outcomes that would otherwise not be possible using Cox models. Therefore, joint models should be used more often in exposure-response analyses of oral targeted anticancer agents.
Collapse
Affiliation(s)
- Lishi Lin
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Vincent van der Noort
- Department of Biometrics, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Gerrina Ruiter
- Department of Thoracic Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Perna G, Pinto E, Spiti A, Torti T, Cucchi M, Caldirola D. Foundations for a Personalized Psycho-Oncology: The State of the Art. J Pers Med 2024; 14:892. [PMID: 39338146 PMCID: PMC11433554 DOI: 10.3390/jpm14090892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Personalized psycho-oncology represents a major challenge for the holistic care of cancer patients. It focuses on individualized psychotherapeutic and psychiatric interventions to address specific psychological needs. This narrative review summarizes the current literature on personalized psycho-oncology and highlights the prevalence and impact of psychiatric/psychological disorders in cancer patients. Personalized approaches, including tailored interventions and interdisciplinary collaboration, have been shown to be effective in improving mental health and overall quality of life. The integration of inflammatory biomarkers into treatment plans is a promising but challenging way to alleviate mental health problems. In addition, there is a need for specific diagnostic tools and treatment guidelines that take into account the specific psychological impact of different types of cancer. Future research should aim to refine these personalized strategies, improve diagnostic accuracy, and evaluate the cost-effectiveness of these interventions to improve both the psychological well-being and treatment outcomes of cancer patients.
Collapse
Affiliation(s)
- Giampaolo Perna
- Department of Biological Sciences, Humanitas University, 20089 Milan, Italy;
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.S.); (M.C.)
| | - Eleonora Pinto
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padua, Italy;
| | - Alessandro Spiti
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.S.); (M.C.)
| | - Tatiana Torti
- ASIPSE School of Cognitive-Behavioral-Therapy, 20124 Milan, Italy;
| | - Michele Cucchi
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy; (A.S.); (M.C.)
| | - Daniela Caldirola
- Department of Biological Sciences, Humanitas University, 20089 Milan, Italy;
| |
Collapse
|
10
|
Al-Shadidi JRMH, Al-Shammari S, Al-Mutairi D, Alkhudhair D, Thu HE, Hussain Z. Chitosan Nanoparticles for Targeted Cancer Therapy: A Review of Stimuli-Responsive, Passive, and Active Targeting Strategies. Int J Nanomedicine 2024; 19:8373-8400. [PMID: 39161363 PMCID: PMC11332424 DOI: 10.2147/ijn.s472433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024] Open
Abstract
Despite all major advancements in drug discovery and development in the pharmaceutical industry, cancer is still one of the most arduous challenges for the scientific community. The implications of nanotechnology have certainly resolved major issues related to conventional anticancer modalities; however, the undesired recognition of nanoparticles (NPs) by the mononuclear phagocyte system (MPS), their poor stability in biological fluids, premature release of payload, and low biocompatibility have restricted their clinical translation. In recent decades, chitosan (CS)-based nanodelivery systems (eg, polymeric NPs, micelles, liposomes, dendrimers, conjugates, solid lipid nanoparticles, etc.) have attained promising recognition from researchers for improving the pharmacokinetics and pharmacodynamics of chemotherapeutics. However, the specialty of this review is to mainly focus on and critically discuss the targeting potential of various CS-based NPs for treatment of different types of cancer. Based on their delivery mechanisms, we classified CS-based NPs into stimuli-responsive, passive, or active targeting nanosystems. Moreover, various functionalization strategies (eg, grafting with polyethylene glycol (PEG), hydrophobic substitution, tethering of stimuli-responsive linkers, and conjugation of targeting ligands) adapted to the architecture of CS-NPs for target-specific delivery of chemotherapeutics have also been considered. Nevertheless, CS-NPs based therapeutics hold great promise for improving therapeutic outcomes while mitigating the off-target effects of chemotherapeutics, a long-term safety profile and clinical testing in humans are warranted for their successful clinical translation.
Collapse
Affiliation(s)
- Jafar R M H Al-Shadidi
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Shahad Al-Shammari
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Danah Al-Mutairi
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Dalal Alkhudhair
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hnin Ei Thu
- Department of Pharmacology, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor Branch, Selangor, Malaysia
| | - Zahid Hussain
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
11
|
Nicholas C, Beharry A, Bendzsak AM, Bisson KR, Dadson K, Dudani S, Iafolla M, Irshad K, Perdrizet K, Raskin W, Singh R, Tsui DCC, Wang X, Yeung C, Cheema PK, Sheffield BS. Point of Care Liquid Biopsy for Cancer Treatment-Early Experience from a Community Center. Cancers (Basel) 2024; 16:2505. [PMID: 39061145 PMCID: PMC11274424 DOI: 10.3390/cancers16142505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Liquid biopsy is rapidly becoming an indispensable tool in the oncologist's arsenal; however, this technique remains elusive in a publicly funded healthcare system, and real-world evidence is needed to demonstrate utility and feasibility. Here, we describe the first experience of an in-house point of care liquid biopsy program at a Canadian community hospital. A retrospective review of consecutive cases that underwent plasma-based next-generation sequencing (NGS) was conducted. Liquid biopsy was initiated at the discretion of clinicians. Sequencing followed a point of care workflow using the Genexus™ integrated sequencer and the Oncomine precision assay, performed by histotechnologists. Results were reported by the attending pathologist. Eligible charts were reviewed for outcomes of interest, including the intent of the liquid biopsy, results of the liquid biopsy, and turnaround time from blood draw to results available. A total of 124 cases, with confirmed or suspected cancer, underwent liquid biopsy between January 2021 and November 2023. The median turnaround time for liquid biopsy results was 3 business days (range 1-12 days). The sensitivity of liquid biopsies was 71%, compared to tissue testing in cases with matched tissue results available for comparison. Common mutations included EGFR (29%), in 86 lung cancer patients, and PIK3CA (22%), identified in 13 breast cancer patients. Healthcare providers ordered liquid biopsies to inform diagnostic investigations and treatment decisions, and to determine progression or resistance mechanisms, as these reasons often overlapped. This study demonstrates that rapid in-house liquid biopsy using point of care methodology is feasible. The technique facilitates precision treatment and offers many additional advantages for cancer care.
Collapse
Affiliation(s)
- Champica Nicholas
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Andrea Beharry
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Anna M. Bendzsak
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Thoracic Surgery, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Kassandra R. Bisson
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Keith Dadson
- Thermo Fisher Scientific, Burlington, ON L7L 5Z1, Canada
| | - Shaan Dudani
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Marco Iafolla
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Kashif Irshad
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Thoracic Surgery, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Kirstin Perdrizet
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - William Raskin
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Raviya Singh
- Division of Medical Oncology, Scarborough Health Network, Scarborough, ON M1P 2V5, Canada
| | - David Chun Cheong Tsui
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Xin Wang
- Division of Medical Oncology, UHN Princess Margaret Cancer Centre, Toronto, ON M5S 1A1, Canada
| | - Ching Yeung
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Thoracic Surgery, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Parneet K. Cheema
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Medical Oncology, William Osler Health System, Brampton, ON L6R 3J7, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Brandon S. Sheffield
- Osler Research Institute for Health Innovation, William Osler Health System, Brampton, ON L6R 3J7, Canada (P.K.C.)
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON L6R 3J7, Canada
| |
Collapse
|
12
|
Forenzo C, Larsen J. Bridging clinical radiotherapy and space radiation therapeutics through reactive oxygen species (ROS)-triggered delivery. Free Radic Biol Med 2024; 219:88-103. [PMID: 38631648 DOI: 10.1016/j.freeradbiomed.2024.04.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
This review explores the convergence of clinical radiotherapy and space radiation therapeutics, focusing on ionizing radiation (IR)-generated reactive oxygen species (ROS). IR, with high-energy particles, induces precise cellular damage, particularly in cancer treatments. The paper discusses parallels between clinical and space IR, highlighting unique characteristics of high-charge and energy particles in space and potential health risks for astronauts. Emphasizing the parallel occurrence of ROS generation in both clinical and space contexts, the review identifies ROS as a crucial factor with dual roles in cellular responses and potential disease initiation. The analysis covers ROS generation mechanisms, variations, and similarities in terrestrial and extraterrestrial environments leading to innovative ROS-responsive delivery systems adaptable for both clinical and space applications. The paper concludes by discussing applications of personalized ROS-triggered therapeutic approaches and discussing the challenges and prospects of implementing these strategies in clinical radiotherapy and extraterrestrial missions. Overall, it underscores the potential of ROS-targeted delivery for advancing therapeutic strategies in terrestrial clinical settings and space exploration, contributing to human health improvement on Earth and beyond.
Collapse
Affiliation(s)
- Chloe Forenzo
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29631, USA
| | - Jessica Larsen
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29631, USA; Department of Bioengineering, Clemson University, Clemson, SC, 29631, USA.
| |
Collapse
|
13
|
Badier L, Quelven I. Zirconium 89 and Copper 64 for ImmunoPET: From Antibody Bioconjugation and Radiolabeling to Molecular Imaging. Pharmaceutics 2024; 16:882. [PMID: 39065579 PMCID: PMC11279968 DOI: 10.3390/pharmaceutics16070882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has transformed cancer treatment. Nevertheless, given the heterogeneity of clinical efficacy, the multiplicity of treatment options available and the possibility of serious adverse effects, selecting the most effective treatment has become the greatest challenge. Molecular imaging offers an attractive way for this purpose. ImmunoPET provides specific imaging with positron emission tomography (PET) using monoclonal antibodies (mAb) or its fragments as vector. By combining the high targeting specificity of mAb and the sensitivity of PET technique, immunoPET could noninvasively and dynamically reveal tumor antigens expression and provide theranostic tools of several types of malignancies. Because of their slow kinetics, mAbs require radioelements defined by a consistent half-life. Zirconium 89 (89Zr) and Copper 64 (64Cu) are radiometals with half-lives suitable for mAb labeling. Radiolabeling with a radiometal requires the prior use of a bifunctional chelate agent (BFCA) to functionalize mAb for radiometal chelation, in a second step. There are a number of BFCA available and much research is focused on antibody functionalization techniques or on developing the optimum chelating agent depending the selected radiometal. In this manuscript, we present a critical account of radiochemical techniques with radionuclides 89Zr and 64Cu and their applications in preclinical and clinical immuno-PET imaging.
Collapse
Affiliation(s)
| | - Isabelle Quelven
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
| |
Collapse
|
14
|
Jóźwiak K, Nguyen VH, Sollfrank L, Linn SC, Hauptmann M. Cox proportional hazards regression in small studies of predictive biomarkers. Sci Rep 2024; 14:14232. [PMID: 38902269 PMCID: PMC11190253 DOI: 10.1038/s41598-024-64573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
Predictive biomarkers are essential for personalized medicine since they select the best treatment for a specific patient. However, of all biomarkers that are evaluated, only few are eventually used in clinical practice. Many promising biomarkers may be erroneously abandoned because they are investigated in small studies using standard statistical techniques which can cause small sample bias or lack of power. The standard technique for failure time endpoints is Cox proportional hazards regression with a multiplicative interaction term between binary variables of biomarker and treatment. Properties of this model in small studies have not been evaluated so far, therefore we performed a simulation study to understand its small sample behavior. As a remedy, we applied a Firth correction to the score function of the Cox model and obtained confidence intervals (CI) using a profile likelihood (PL) approach. These methods are generally recommended for small studies of different design. Our results show that a Cox model estimates the biomarker-treatment interaction term and the treatment effect in one of the biomarker subgroups with bias, and overestimates their standard errors. Bias is however reduced and power is increased with Firth correction and PL CIs. Hence, the modified Cox model and PL CI should be used instead of a standard Cox model with Wald based CI in small studies of predictive biomarkers.
Collapse
Affiliation(s)
- K Jóźwiak
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Fehrbelliner Straße 39, 16816, Neuruppin, Germany.
| | - V H Nguyen
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Fehrbelliner Straße 39, 16816, Neuruppin, Germany
- Leibniz Centre for Agricultural Landscape Research (ZALF), Müncheberg, Germany
| | - L Sollfrank
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Fehrbelliner Straße 39, 16816, Neuruppin, Germany
| | - S C Linn
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center, Utrecht, The Netherlands
| | - M Hauptmann
- Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Fehrbelliner Straße 39, 16816, Neuruppin, Germany
| |
Collapse
|
15
|
Aslan H, Renzi G, Angeli A, D'Agostino I, Ronca R, Massardi ML, Tavani C, Carradori S, Ferraroni M, Governa P, Manetti F, Carta F, Supuran CT. Benzenesulfonamide decorated dihydropyrimidin(thi)ones: carbonic anhydrase profiling and antiproliferative activity. RSC Med Chem 2024; 15:1929-1941. [PMID: 38911163 PMCID: PMC11187566 DOI: 10.1039/d4md00101j] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/24/2024] [Indexed: 06/25/2024] Open
Abstract
In the last decades, carbonic anhydrases (CAs) have become the top investigated innovative pharmacological targets and, in particular, isoforms IX and XII have been widely studied due to the evidence of their overexpression in hypoxic tumors. The frantic race to find new anticancer agents places the quick preparation of large libraries of putative bioactive compounds as the basis of a successful drug discovery and development programme. In this context, multi-component and, in general, one-step reactions are becoming very popular and, among them, Biginelli's reaction gave clean and easy-to-isolate products. Thus, we synthesized a series of Biginelli's products (10-17a-b) and similar derivatives (20-21) bearing the benzenesulfonamide moiety, which is known to inhibit CA enzymes. Through the stopped-flow technique, we were able to assess their ability to inhibit the targeted CAs IX and XII in the nanomolar range with promising selectivity over the physiologically relevant isoforms I and II. Crystallography studies and docking simulations helped us to gain insight into the interaction patterns established in the enzyme-inhibitor complex. From a chemical similarity-based screening of in-house libraries of compounds, a diphenylpyrimidine (23) emerged. The surprisingly potent inhibitory activity of 23 for CAs IX and XII along with its strong antiproliferative effect on two (triple-negative breast cancer MDA-MB-231 and glioblastoma U87MG) cell lines laid the foundation for further investigation, again confirming the key role of CAs in cancer.
Collapse
Affiliation(s)
- Hakan Aslan
- Department of Chemistry, Faculty of Science and Arts, Sinop University Sinop Turkey
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence Sesto Fiorentino Florence 50019 Italy
| | - Gioele Renzi
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence Sesto Fiorentino Florence 50019 Italy
| | - Andrea Angeli
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence Sesto Fiorentino Florence 50019 Italy
| | - Ilaria D'Agostino
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence Sesto Fiorentino Florence 50019 Italy
- Department of Pharmacy, University of Pisa 56126 Pisa Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia 25123 Brescia Italy
| | - Maria Luisa Massardi
- Department of Molecular and Translational Medicine, University of Brescia 25123 Brescia Italy
| | - Camilla Tavani
- Department of Molecular and Translational Medicine, University of Brescia 25123 Brescia Italy
| | - Simone Carradori
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara 66100 Chieti Italy
| | - Marta Ferraroni
- Department of Chemistry "Ugo Schiff", University of Florence Sesto Fiorentino Florence 50019 Italy
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena 53100 Siena Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena 53100 Siena Italy
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence Sesto Fiorentino Florence 50019 Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence Sesto Fiorentino Florence 50019 Italy
| |
Collapse
|
16
|
Arnold CR, Mangesius J, Portnaia I, Ganswindt U, Wolff HA. Innovative therapeutic strategies to overcome radioresistance in breast cancer. Front Oncol 2024; 14:1379986. [PMID: 38873260 PMCID: PMC11169591 DOI: 10.3389/fonc.2024.1379986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/10/2024] [Indexed: 06/15/2024] Open
Abstract
Despite a comparatively favorable prognosis relative to other malignancies, breast cancer continues to significantly impact women's health globally, partly due to its high incidence rate. A critical factor in treatment failure is radiation resistance - the capacity of tumor cells to withstand high doses of ionizing radiation. Advancements in understanding the cellular and molecular mechanisms underlying radioresistance, coupled with enhanced characterization of radioresistant cell clones, are paving the way for the development of novel treatment modalities that hold potential for future clinical application. In the context of combating radioresistance in breast cancer, potential targets of interest include long non-coding RNAs (lncRNAs), micro RNAs (miRNAs), and their associated signaling pathways, along with other signal transduction routes amenable to pharmacological intervention. Furthermore, technical, and methodological innovations, such as the integration of hyperthermia or nanoparticles with radiotherapy, have the potential to enhance treatment responses in patients with radioresistant breast cancer. This review endeavors to provide a comprehensive survey of the current scientific landscape, focusing on novel therapeutic advancements specifically addressing radioresistant breast cancer.
Collapse
Affiliation(s)
| | - Julian Mangesius
- Department of Radiation-Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Iana Portnaia
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Ute Ganswindt
- Department of Radiation-Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hendrik Andreas Wolff
- Department of Radiology, Nuclear Medicine, and Radiotherapy, Radiology Munich, Munich, Germany
| |
Collapse
|
17
|
El-Tanani M, Rabbani SA, Babiker R, Rangraze I, Kapre S, Palakurthi SS, Alnuqaydan AM, Aljabali AA, Rizzo M, El-Tanani Y, Tambuwala MM. Unraveling the tumor microenvironment: Insights into cancer metastasis and therapeutic strategies. Cancer Lett 2024; 591:216894. [PMID: 38626856 DOI: 10.1016/j.canlet.2024.216894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 05/03/2024]
Abstract
This comprehensive review delves into the pivotal role of the tumor microenvironment (TME) in cancer metastasis and therapeutic response, offering fresh insights into the intricate interplay between cancer cells and their surrounding milieu. The TME, a dynamic ecosystem comprising diverse cellular and acellular elements, not only fosters tumor progression but also profoundly affects the efficacy of conventional and emerging cancer therapies. Through nuanced exploration, this review illuminates the multifaceted nature of the TME, elucidating its capacity to engender drug resistance via mechanisms such as hypoxia, immune evasion, and the establishment of physical barriers to drug delivery. Moreover, it investigates innovative therapeutic approaches aimed at targeting the TME, including stromal reprogramming, immune microenvironment modulation, extracellular matrix (ECM)-targeting agents, and personalized medicine strategies, highlighting their potential to augment treatment outcomes. Furthermore, this review critically evaluates the challenges posed by the complexity and heterogeneity of the TME, which contribute to variable therapeutic responses and potentially unintended consequences. This underscores the need to identify robust biomarkers and advance predictive models to anticipate treatment outcomes, as well as advocate for combination therapies that address multiple facets of the TME. Finally, the review emphasizes the necessity of an interdisciplinary approach and the integration of cutting-edge technologies to unravel the intricacies of the TME, thereby facilitating the development of more effective, adaptable, and personalized cancer treatments. By providing critical insights into the current state of TME research and its implications for the future of oncology, this review highlights the dynamic and evolving landscape of this field.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Syed Arman Rabbani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Rasha Babiker
- Physiology Department, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras-al-Khaimah, United Arab Emirates
| | - Imran Rangraze
- Internal Medicine Department, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras-al-Khaimah, United Arab Emirates
| | - Sumedha Kapre
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX, 78363, USA
| | - Sushesh Srivastsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX, 78363, USA
| | - Abdullah M Alnuqaydan
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia.
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, 21163, Jordan
| | - Manfredi Rizzo
- (D)epartment of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, Palermo, Italy
| | - Yahia El-Tanani
- Medical School, St George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE, UK.
| | - Murtaza M Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates; Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
| |
Collapse
|
18
|
Zhang J, Zhu W, Ma Y, Huang X, Su W, Sun Y, Liu Q, Ma T, Ma L, Sun J, Fan S, Wang X, Lin S, Wang W, Han C. Triphenylphosphonium-linked derivative of hecogenin with enhanced antiproliferative activity: Design, synthesis, and biological evaluation. Bioorg Chem 2024; 145:107210. [PMID: 38364551 DOI: 10.1016/j.bioorg.2024.107210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/29/2024] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
Hecogenin (HCG), a steroidal sapogenin, possesses good antitumor properties. However, the application of HCG for cancer treatment has been hindered primarily by its moderate potency. In this study, we incorporated triphenylphosphonium cation (TPP+) at the C-3 and C-12 positions through different lengths of alkyl chains to target mitochondria and enhance the efficacy and selectivity of the parent compound. Cytotoxicity screening revealed that most of the target compounds exhibited potent antiproliferative activity against five human cancer cell lines (MKN45, A549, HCT-116, MCF-7, and HepG2). Structure-activity relationship studies indicated that the TPP+ group significantly enhanced the antiproliferative potency of HCG. Among these compounds, 3c demonstrated remarkable potency against MKN45 cells with an IC50 value of 0.48 μM, significantly more effective than its parent compound HCG (IC50 > 100 μM). Further investigations into the mechanism of action revealed that 3c induced apoptosis of MKN45 cells through the mitochondrial pathway. In a zebrafish xenograft model, 3c inhibited the proliferation of MKN45 cells. Overall, these results suggest that 3c, with potent antiproliferative activity, may serve as a valuable scaffold for developing new antitumor agents.
Collapse
Affiliation(s)
- Jinling Zhang
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Wenquan Zhu
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Yukun Ma
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Xiaoying Huang
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Wenle Su
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Yu Sun
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Qi Liu
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Tiancheng Ma
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Liwei Ma
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Jia Sun
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Songjie Fan
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Xiaoli Wang
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Song Lin
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Wenbao Wang
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China.
| | - Cuiyan Han
- Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China.
| |
Collapse
|
19
|
Kreidieh F, McQuade J. Novel insights into cardiovascular toxicity of cancer targeted and immune therapies: Beyond ischemia with non-obstructive coronary arteries (INOCA). AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 40:100374. [PMID: 38510501 PMCID: PMC10946000 DOI: 10.1016/j.ahjo.2024.100374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Novel immune and targeted therapies approved over the past 2 decades have resulted in dramatic improvements in cancer-specific outcomes for many cancer patients. However, many of these agents can induce cardiovascular toxicity in a subset of patients. The field of cardio-oncology was established based on observations that anti-neoplastic chemotherapies and mantle radiation can lead to premature cardiomyopathy in cancer survivors. While conventional chemotherapy, targeted therapy, and immune therapies can all result in cardiovascular adverse events, the mechanisms, timing, and incidence of these events are inherently different. Many of these effects converge upon the coronary microvasculature to involve, through endocardial endothelial cells, a more direct effect through close proximity to cardiomyocyte with cellular communication and signaling pathways. In this review, we will provide an overview of emerging paradigms in the field of Cardio-Oncology, particularly the role of the coronary microvasculature in mediating cardiovascular toxicity of important cancer targeted and immune therapies. As the number of cancer patients treated with novel immune and targeted therapies grows exponentially and subsequently the number of long-term cancer survivors dramatically increases, it is critical that cardiologists and cardiology researchers recognize the unique potential cardiovascular toxicities of these agents.
Collapse
Affiliation(s)
- Firas Kreidieh
- Instructor of Clinical Medicine- Division of Hematology-Oncology; Associate Director- Internal Medicine Residency Program, American University of Beirut, Beirut, Lebanon
| | - Jennifer McQuade
- Associate Professor and Physician Scientist in Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
20
|
Hoshino A, Oana Y, Ohi Y, Maeda Y, Omori M, Takada Y, Ikeda T, Sotome K, Maeda H, Yanagisawa T, Takeuchi O, Kuronuma S, Sangai T, Shibahara Y, Murakumo Y, Saegusa M, Kanomata N, Nagasawa S, Yamaguchi R, Yoshida M, Kozuka Y, Matsumoto H, Tsugawa K, Maeda I. Using the DNA Integrity Number to Analyze DNA Quality in Specimens Collected from Liquid-Based Cytology after Fine-Needle Aspiration of Breast Tumors and Lesions. Acta Cytol 2024; 68:145-152. [PMID: 38555634 DOI: 10.1159/000538071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/25/2024] [Indexed: 04/02/2024]
Abstract
INTRODUCTION Cancer genome analysis using next-generation sequencing requires adequate and high-quality DNA samples. Genomic analyses were conventionally performed using formalin-fixed paraffin-embedded sections rather than cytology samples such as cell block or smear specimens. Specimens collected from liquid-based cytology (LBC) have the potential to be sources of high-quality DNA suitable for genetic analysis even after long-term storage. METHODS We collected breast tumor/lesion fractions from 92 residual LBC specimens using fine-needle aspiration (FNA) biopsy, including breast carcinoma (1 invasive carcinoma and 4 ductal carcinomas in situ), papillomatous lesion (5 intraductal papillomas), and fibroepithelial lesion (19 phyllodes tumors and 53 fibroadenomas) samples, and others (1 ductal adenoma, 1 hamartoma, 1 fibrocystic disease, and 7 unknown). DNA was extracted from all samples and subjected to DNA integrity number (DIN) score analysis. RESULTS Average DIN score collected from 92 LBC specimens was significantly higher score. In addition, high-quality DNA with high DIN values (7.39 ± 0.80) was successfully extracted more than 12 months after storage of residual LBC specimens. CONCLUSION Residual LBC specimens collected from FNA of the breast were verified to carry high-quality DNA and could serve as an alternate source for genetic analysis.
Collapse
Affiliation(s)
- Akiyoshi Hoshino
- Department of Diagnostic Pathology, Kitasato University Kitasato Institute Hospital, Tokyo, Japan,
- Department of Pathology, Kitasato University School of Medicine, Tokyo, Japan,
| | - Yoshiyasu Oana
- Department of Diagnostic Pathology, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Yasuyo Ohi
- Department of Pathology, Sagara Hospital, Kagoshima City, Kagoshima, Japan
| | - Yukari Maeda
- Department of Pathology, Sagara Hospital, Kagoshima City, Kagoshima, Japan
| | - Masako Omori
- Department of Pathology, Kurashiki Medical Center, Kurashiki City, Okayama, Japan
| | - Yuki Takada
- Department of Pathology, Kurashiki Medical Center, Kurashiki City, Okayama, Japan
| | - Tadashi Ikeda
- Department of Surgery, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Keiichi Sotome
- Department of Surgery, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Hinako Maeda
- Department of Surgery, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Takako Yanagisawa
- Department of Surgery, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Osamu Takeuchi
- Biomedical Laboratory, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Satoshi Kuronuma
- Biomedical Laboratory, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Takafumi Sangai
- Department of Surgery, Kitasato University Hospital, Sagamihara City, Sagamihara, Japan
| | - Yukiko Shibahara
- Department of Pathology, Kitasato University Hospital, Sagamihara City, Sagamihara, Japan
| | - Yoshiki Murakumo
- Department of Pathology, Kitasato University Hospital, Sagamihara City, Sagamihara, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University Hospital, Sagamihara City, Sagamihara, Japan
| | - Naoki Kanomata
- Department of Pathology, St. Lukes International Hospital, Tokyo, Japan
| | - Satoi Nagasawa
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa City, Chiba, Japan
| | - Rin Yamaguchi
- Department of Pathology, Nagasaki University Hospital, Nagasaki City, Nagasaki, Japan
| | - Masayuki Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuji Kozuka
- Department of Pathology, Mie University Hospital, Tsu, Japan
| | | | - Koichiro Tsugawa
- Divison of Breast and Endocrine Surgery, Department of Surgery, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ichiro Maeda
- Department of Diagnostic Pathology, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
- Department of Pathology, Kitasato University School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Welsh AM, Muljo SA. Post-transcriptional (re)programming of B lymphocyte development: From bench to bedside? Adv Immunol 2024; 161:85-108. [PMID: 38763703 DOI: 10.1016/bs.ai.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Hematopoiesis, a process which generates blood and immune cells, changes significantly during mammalian development. Definitive hematopoiesis is marked by the emergence of long-term hematopoietic stem cells (HSCs). Here, we will focus on the post-transcriptional differences between fetal liver (FL) and adult bone marrow (ABM) HSCs. It remains unclear how or why exactly FL HSCs transition to ABM HSCs, but we aim to leverage their differences to revive an old idea: in utero HSC transplantation. Unexpectedly, the expression of certain RNA-binding proteins (RBPs) play an important role in HSC specification, and can be employed to convert or reprogram adult HSCs back to a fetal-like state. Among other features, FL HSCs have a broad differentiation capacity that includes the ability to regenerate both conventional B and T cells, as well as innate-like or unconventional lymphocytes such as B-1a and marginal zone B (MzB) cells. This chapter will focus on RNA binding proteins, namely LIN28B and IGF2BP3, that are expressed during fetal life and how they promote B-1a cell development. Furthermore, this chapter considers a potential clinical application of synthetic co-expression of LIN28B and IGF2BP3 in HSCs.
Collapse
Affiliation(s)
- Alia M Welsh
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stefan A Muljo
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States.
| |
Collapse
|
22
|
Schroeder J, Lagisetty K, Lynch W, Lin J, Chang AC, Reddy RM. Rural Women Have a Prolonged Recovery Process after Esophagectomy. Cancers (Basel) 2024; 16:1078. [PMID: 38539414 PMCID: PMC10968561 DOI: 10.3390/cancers16061078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND Gender and geographic access to care play a large role in health disparities in esophageal cancer care. The aim of our study was to evaluate disparities in peri-operative outcomes for patients undergoing esophagectomy based on gender and geographic location. METHODS A retrospective cohort of prospectively collected data from patients who underwent esophagectomy from 2003 to 2022 was identified and analyzed based on gender and county, which were aggregated into existing state-level "metropolitan" versus "rural" designations. The demographics, pre-operative treatment, surgical complications, post-operative outcomes, and length of stay (LOS) of each group were analyzed using chi-squared, paired t-tests and single-factor ANOVA. RESULTS Of the 1545 patients, men (83.6%) and women (16.4%) experienced similar rates of post-operative complications, but women experienced significantly longer hospital (p = 0.002) and ICU (p = 0.03) LOSs as compared with their male counterparts, with no differences in 30-day mortality. When separated by geographic criteria, rural women were further outliers, with significantly longer hospital LOSs (p < 0.001) and higher rates of ICU admission (p < 0.001). CONCLUSIONS Rural female patients undergoing esophagectomy were more likely to have a longer inpatient recovery process compared with their female metropolitan or male counterparts, suggesting a need for more targeted interventions in this population.
Collapse
Affiliation(s)
- Julia Schroeder
- University of Michigan Medical School, 3808 Medical Science Bldg, Ann Arbor, MI 48109, USA
| | - Kiran Lagisetty
- University of Michigan Medical School, 3808 Medical Science Bldg, Ann Arbor, MI 48109, USA
- Michigan Medicine, Section of Thoracic Surgery, Department of Surgery, 1500 E. Medical Center Drive, TC 2120, Ann Arbor, MI 48109, USA
| | - William Lynch
- University of Michigan Medical School, 3808 Medical Science Bldg, Ann Arbor, MI 48109, USA
- Michigan Medicine, Section of Thoracic Surgery, Department of Surgery, 1500 E. Medical Center Drive, TC 2120, Ann Arbor, MI 48109, USA
| | - Jules Lin
- University of Michigan Medical School, 3808 Medical Science Bldg, Ann Arbor, MI 48109, USA
- Michigan Medicine, Section of Thoracic Surgery, Department of Surgery, 1500 E. Medical Center Drive, TC 2120, Ann Arbor, MI 48109, USA
| | - Andrew C. Chang
- University of Michigan Medical School, 3808 Medical Science Bldg, Ann Arbor, MI 48109, USA
- Michigan Medicine, Section of Thoracic Surgery, Department of Surgery, 1500 E. Medical Center Drive, TC 2120, Ann Arbor, MI 48109, USA
| | - Rishindra M. Reddy
- University of Michigan Medical School, 3808 Medical Science Bldg, Ann Arbor, MI 48109, USA
- Michigan Medicine, Section of Thoracic Surgery, Department of Surgery, 1500 E. Medical Center Drive, TC 2120, Ann Arbor, MI 48109, USA
| |
Collapse
|
23
|
Kim S, Lee Y, Song BR, Sim H, Kang EH, Hwang M, Yu N, Hong S, Park C, Ahn BC, Lim EJ, Hwang KH, Park SY, Choi JH, Lee GK, Han JY. Drug Response of Patient-Derived Lung Cancer Cells Predicts Clinical Outcomes of Targeted Therapy. Cancers (Basel) 2024; 16:778. [PMID: 38398169 PMCID: PMC10887363 DOI: 10.3390/cancers16040778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Intratumor heterogeneity leads to different responses to targeted therapies, even within patients whose tumors harbor identical driver oncogenes. This study examined clinical outcomes according to a patient-derived cell (PDC)-based drug sensitivity test in lung cancer patients treated with targeted therapies. From 487 lung cancers, 397 PDCs were established with a success rate of 82%. In 139 PDCs from advanced non-small-cell lung cancer (NSCLC) patients receiving targeted therapies, the standardized area under the curve (AUC) values for the drugs was significantly correlated with their tumor response (p = 0.002). Among 59 chemo-naive EGFR/ALK-positive NSCLC patients, the PDC non-responders showed a significantly inferior response rate (RR) and progression-free survival (PFS) for the targeted drugs than the PDC responders (RR, 25% vs. 78%, p = 0.011; median PFS, 3.4 months [95% confidence interval (CI), 2.8-4.1] vs. 11.8 months [95% CI, 6.5-17.0], p < 0.001). Of 25 EGFR-positive NSCLC patients re-challenged with EGFR inhibitors, the PDC responder showed a higher RR than the PDC non-responder (42% vs. 15%). Four patients with wild-type EGFR or uncommon EGFR-mutant NSCLC were treated with EGFR inhibitors based on their favorable PDC response to EGFR inhibitors, and two patients showed dramatic responses. Therefore, the PDC-based drug sensitivity test results were significantly associated with clinical outcomes in patients with EGFR- or ALK-positive NSCLC. It may be helpful for predicting individual heterogenous clinical outcomes beyond genomic alterations.
Collapse
Affiliation(s)
- Sunshin Kim
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Youngjoo Lee
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
- Center for Lung Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (B.-C.A.); (E.J.L.); (K.H.H.); (J.-H.C.)
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center, Goyang 10408, Republic of Korea
| | - Bo Ram Song
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Hanna Sim
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Eun Hye Kang
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Mihwa Hwang
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Namhee Yu
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Sehwa Hong
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Charny Park
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
| | - Beung-Chul Ahn
- Center for Lung Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (B.-C.A.); (E.J.L.); (K.H.H.); (J.-H.C.)
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center, Goyang 10408, Republic of Korea
| | - Eun Jin Lim
- Center for Lung Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (B.-C.A.); (E.J.L.); (K.H.H.); (J.-H.C.)
| | - Kum Hui Hwang
- Center for Lung Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (B.-C.A.); (E.J.L.); (K.H.H.); (J.-H.C.)
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center, Goyang 10408, Republic of Korea; (S.-Y.P.); (G.K.L.)
| | - Jin-Ho Choi
- Center for Lung Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (B.-C.A.); (E.J.L.); (K.H.H.); (J.-H.C.)
| | - Geon Kook Lee
- Department of Pathology, National Cancer Center, Goyang 10408, Republic of Korea; (S.-Y.P.); (G.K.L.)
| | - Ji-Youn Han
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (S.K.); (Y.L.); (B.R.S.); (H.S.); (E.H.K.); (M.H.); (N.Y.); (S.H.); (C.P.)
- Center for Lung Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (B.-C.A.); (E.J.L.); (K.H.H.); (J.-H.C.)
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center, Goyang 10408, Republic of Korea
| |
Collapse
|
24
|
Farooq A, Hassan M, Loya A, Asghar K. Community Outreach and Engagement in Cancer Research Through a Biobank Clinic at Shaukat Khanum Memorial Cancer Hospital and Research Centre, Pakistan. Cureus 2024; 16:e55179. [PMID: 38558595 PMCID: PMC10980601 DOI: 10.7759/cureus.55179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Cancer's increasing prevalence across the globe emphasizes the urgency for continued research, prevention, and accessible healthcare to mitigate its impact on individuals and communities. While there have been significant advances made towards controlling cancer morbidity and mortality in recent decades, Pakistan continues to experience a markedly elevated burden of the disease. With this study, we aim to raise awareness about biobank research within the cancer patient community, fostering participation and collaboration to advance the fight against cancer through vital research contributions. METHODS In October 2022, we initiated the biobank clinic at Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC). Here, patients underwent screening and received invitations to voluntarily participate in biobank research. During these interactions, we engaged patients in discussions about the significance of biobank research, addressed their concerns, and encouraged their participation in advancing our research endeavors. Two-sample independent t-tests were performed to compare the mean number of participants in pre-clinic and post-clinic cohorts. RESULTS This research involved a total of 958 participants, with 312 participants enrolled before the clinic and 646 participants enrolled after the clinic. We have observed a noticeable increase in the participation of cancer patients in our research endeavors since the inception of the biobank clinic (p-value<0.001). Over an 11-month time frame, we scheduled appointments for 759 patients, and out of those, 656 patients availed themselves to visit the clinic. Impressively, we achieved the enrollment of 646 patients into the clinic, reflecting an exceptional consent rate of 98.47% for their active involvement in our research initiatives. This underscores our commitment to conducting comprehensive discussions and providing thorough explanations regarding the ethical and procedural aspects of our research. CONCLUSION Biobank clinic plays a pivotal role in raising cancer awareness and fostering research participation, especially in regions with limited healthcare infrastructure and lower literacy rates. It emerges as a community-engagement model that aligns research with local needs, ensuring its relevance and benefit to the population.
Collapse
Affiliation(s)
- Asim Farooq
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Muhammad Hassan
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Asif Loya
- Pathology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Kashif Asghar
- Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| |
Collapse
|
25
|
Bui NL, Chu DT. An introduction to RNA therapeutics and their potentials. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 203:1-12. [PMID: 38359993 DOI: 10.1016/bs.pmbts.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
RNA therapeutics is a biological term regarding the usage of RNA-based molecules for medical purposes. Thanks to the success of mRNA-vaccine production against COVID-19, RNA therapeutics has gained more and more attention and investigation from worldwide scientists. It is considered as one of the promising alternatives for conventional drugs. In this first chapter, we presented an overview of the history and perspectives of RNA therapeutics' development. This chapter also explained the underlying mechanisms of different RNA-based molecules, including antisense oligonucleotide, interfering RNA (iRNA), aptamer, and mRNA, from degrading mRNA to inactivating targeted protein. Although there are many advantages of RNA therapeutics, its challenges in designing RNA chemical structure and the delivery vehicle need to be discussed. We described advanced technologies in the development of drug delivery systems that are positively correlated to the efficacy of the drug. Our aim is to provide a general background of RNA therapeutics to the audience before introducing plenty of more detailed parts, including clinical applications in certain diseases in the following chapters of the "RNA therapeutics" book.
Collapse
Affiliation(s)
- Nhat-Le Bui
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam.
| |
Collapse
|
26
|
Khan SI, Hassan A, Bano R, Gilani MA, Marty JL, Zhang H, Hayat A. An innovative and universal dual-signal ratiometric spectro-electrochemical imprinted sensor design for sandwich type detection of anticancer-drug, gemcitabine, in serum samples; cross validation via computational modeling. Talanta 2024; 267:125233. [PMID: 37774453 DOI: 10.1016/j.talanta.2023.125233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
An innovative and universal imprinted sensor design for sandwich type detection of gemcitabine (GMT) in human serum samples is described. GMT is widely used in the treatment of different tumors, such as lung, ovarian, pancreatic, and breast cancer. The serum albumin-drug interaction was translated to design a multifunctional, ratiometric and dual mode silver nanoparticle based probe (BSA-Ag nanoprobe), as a read out system. Subsequently, polypyrrol imprinted drug receptor sites was engineered to selectively capture the GMT on the transducer surface. The GMT was sandwiched between imprinted receptor surface and BSA-Ag nanoprobe to generate the spectro-electrochemical signals. The formation of nanoprobe was confirmed through various characterization techniques, including X-ray diffraction (XRD), Fourier transform infrared (FTIR) spectroscopy, micro-Raman spectroscopy, Dynamic light scattering (DLS), and UV-Visible (UV-Vis) analysis, while each step of sensor fabrication was characterized via field emission scanning electron microscope (FE-SEM), Static water Contact angle measurements, cyclic voltammetry (CV), and electrochemical impedance spectroscopy (EIS). Different variable parameters were optimized to improve the analytical performance of the sensor design. Under optimal conditions, spectro-electrochemical sensor permitted linear ranges between 1 and 200 μmol L-1 and 0.5-200 μmol L-1, with limits of detection (LOD) of 0.4 μmol L-1 and 0.15 μmol L-1 respectively. Furthermore, the designed sensor successfully differentiated the serum samples of lung cancer patients and healthy volunteers. The obtained results were validated with standard Liquid chromatography-mass spectrometry (LC/MS) analysis of the patients and healthy volunteer's serum samples. Lastly, density functional theory (DFT) and molecular docking calculations revealed the enhanced GMT binding capability of molecularly imprinted polypyrrole and molecular level interaction between the GMT and BSA, to validate the sandwich sensor design.
Collapse
Affiliation(s)
- Shaista Ijaz Khan
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, PO Box 250353, Jinan, Shandong, China; Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSAT University Islamabad, Lahore Campus, 1.5 KM Defence Road Off Raiwind Road, Lahore, Pakistan
| | - Ayaz Hassan
- Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSAT University Islamabad, Lahore Campus, 1.5 KM Defence Road Off Raiwind Road, Lahore, Pakistan
| | - Rehana Bano
- Department of Chemistry, COMSATS University Islamabad, Lahore Campus, 1.5 KM Defence Road Off Raiwind Road, Lahore, Pakistan
| | - Mazhar Amjad Gilani
- Department of Chemistry, COMSATS University Islamabad, Lahore Campus, 1.5 KM Defence Road Off Raiwind Road, Lahore, Pakistan
| | | | - Hongxia Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, PO Box 250353, Jinan, Shandong, China.
| | - Akhtar Hayat
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, PO Box 250353, Jinan, Shandong, China; Interdisciplinary Research Center in Biomedical Materials (IRCBM), COMSAT University Islamabad, Lahore Campus, 1.5 KM Defence Road Off Raiwind Road, Lahore, Pakistan.
| |
Collapse
|
27
|
Saikia S, Postwala H, Athilingam VP, Anandan A, Padma VV, Kalita PP, Chorawala M, Prajapati B. Single Nucleotide Polymorphisms (SNPs) in the Shadows: Uncovering their Function in Non-Coding Region of Esophageal Cancer. Curr Pharm Biotechnol 2024; 25:1915-1938. [PMID: 38310451 DOI: 10.2174/0113892010265004231116092802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 02/05/2024]
Abstract
Esophageal cancer is a complex disease influenced by genetic and environmental factors. Single nucleotide polymorphisms (SNPs) in non-coding regions of the genome have emerged as crucial contributors to esophageal cancer susceptibility. This review provides a comprehensive overview of the role of SNPs in non-coding regions and their association with esophageal cancer. The accumulation of SNPs in the genome has been implicated in esophageal cancer risk. Various studies have identified specific locations in the genome where SNPs are more likely to occur, suggesting a location-specific response. Chromatin conformational studies have shed light on the localization of SNPs and their impact on gene transcription, posttranscriptional modifications, gene expression regulation, and histone modification. Furthermore, miRNA-related SNPs have been found to play a significant role in esophageal squamous cell carcinoma (ESCC). These SNPs can affect miRNA binding sites, thereby altering target gene regulation and contributing to ESCC development. Additionally, the risk of ESCC has been linked to base excision repair, suggesting that SNPs in this pathway may influence disease susceptibility. Somatic DNA segment alterations and modified expression quantitative trait loci (eQTL) have also been associated with ESCC. These alterations can lead to disrupted gene expression and cellular processes, ultimately contributing to cancer development and progression. Moreover, SNPs have been found to be associated with the long non-coding RNA HOTAIR, which plays a crucial role in ESCC pathogenesis. This review concludes with a discussion of the current and future perspectives in the field of SNPs in non-coding regions and their relevance to esophageal cancer. Understanding the functional implications of these SNPs may lead to the identification of novel therapeutic targets and the development of personalized approaches for esophageal cancer prevention and treatment.
Collapse
Affiliation(s)
- Surovi Saikia
- Department of Natural Product Chemistry, Translational Research Laboratory, Bharathiar University, Coimbatore - 641 046, Tamil Nadu, India
| | - Humzah Postwala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, India
| | - Vishnu Prabhu Athilingam
- Department of Natural Product Chemistry, Translational Research Laboratory, Bharathiar University, Coimbatore - 641 046, Tamil Nadu, India
| | - Aparna Anandan
- Department of Natural Product Chemistry, Translational Research Laboratory, Bharathiar University, Coimbatore - 641 046, Tamil Nadu, India
| | - V Vijaya Padma
- Department of Natural Product Chemistry, Translational Research Laboratory, Bharathiar University, Coimbatore - 641 046, Tamil Nadu, India
| | - Partha P Kalita
- Program of Biotechnology, Assam Down Town University, Panikhaiti, Guwahati 781026, Assam, India
| | - Mehul Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, India
| | - Bhupendra Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree. S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat, India
| |
Collapse
|
28
|
Zehra M, Fatima T, Hanif A, Raufi N, Khan A. Nadofaragene: a new era of precision medicine for bladder cancer. Ann Med Surg (Lond) 2024; 86:7-10. [PMID: 38222686 PMCID: PMC10783406 DOI: 10.1097/ms9.0000000000001488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/30/2023] [Indexed: 01/16/2024] Open
Affiliation(s)
- Maha Zehra
- Department of Medicine, Dow University of Health Sciences
| | - Tehreem Fatima
- Department of Medicine, Dow University of Health Sciences
| | - Areeba Hanif
- Dow University of Health Sciences, Karachi, Pakistan
| | - Nahid Raufi
- Department of Medicine, Kabul Medical University, Afghanistan
| | - Afsheen Khan
- Department of Medicine, Dow University of Health Sciences
| |
Collapse
|
29
|
Shams M, Abdallah S, Alsadoun L, Hamid YH, Gasim R, Hassan A. Oncological Horizons: The Synergy of Medical and Surgical Innovations in Cancer Treatment. Cureus 2023; 15:e49249. [PMID: 38143618 PMCID: PMC10743204 DOI: 10.7759/cureus.49249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2023] [Indexed: 12/26/2023] Open
Abstract
The landscape of cancer treatment has witnessed a remarkable transformation in recent years, marked by the convergence of medical and surgical innovations. Historically, cancer therapy faced challenges, including limited efficacy and severe side effects. This narrative review explores the historical progression of cancer treatments, shedding light on significant breakthroughs in both medical and surgical oncology. It comprehensively addresses the medical domain, covering chemotherapy, targeted therapies, immunotherapy, hormonal treatments, and radiological procedures. Simultaneously, it delves into the surgical realm, discussing the evolution of surgical techniques, minimally invasive procedures, and the role of surgery across various stages of cancer. The article emphasizes the fusion of medical and surgical approaches, highlighting neoadjuvant and adjuvant therapies and the significance of multidisciplinary tumor boards. It also addresses innovations, challenges, and the pivotal role of patient-centered care. Furthermore, it offers insights into the future directions and forecasts in the constantly evolving field of integrated oncological care. This review provides a comprehensive understanding of the dynamic and transformative nature of cancer treatment, reflecting the unwavering commitment of the medical and surgical communities in the ongoing fight against cancer.
Collapse
Affiliation(s)
| | | | - Lara Alsadoun
- Trauma and Orthopaedics, Chelsea and Westminster Hospital, London, GBR
| | - Yusra H Hamid
- Community Medicine, Faculty of Medicine, University of Khartoum, Khartoum, SDN
| | - Rayan Gasim
- Internal Medicine, University of Khartoum, Khartoum, SDN
| | | |
Collapse
|
30
|
Bhattacharyya S, Ehsan SF, Karacosta LG. Phenotypic maps for precision medicine: a promising systems biology tool for assessing therapy response and resistance at a personalized level. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1256104. [PMID: 37964768 PMCID: PMC10642209 DOI: 10.3389/fnetp.2023.1256104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/28/2023] [Indexed: 11/16/2023]
Abstract
In this perspective we discuss how tumor heterogeneity and therapy resistance necessitate a focus on more personalized approaches, prompting a shift toward precision medicine. At the heart of the shift towards personalized medicine, omics-driven systems biology becomes a driving force as it leverages high-throughput technologies and novel bioinformatics tools. These enable the creation of systems-based maps, providing a comprehensive view of individual tumor's functional plasticity. We highlight the innovative PHENOSTAMP program, which leverages high-dimensional data to construct a visually intuitive and user-friendly map. This map was created to encapsulate complex transitional states in cancer cells, such as Epithelial-Mesenchymal Transition (EMT) and Mesenchymal-Epithelial Transition (MET), offering a visually intuitive way to understand disease progression and therapeutic responses at single-cell resolution in relation to EMT-related single-cell phenotypes. Most importantly, PHENOSTAMP functions as a reference map, which allows researchers and clinicians to assess one clinical specimen at a time in relation to their phenotypic heterogeneity, setting the foundation on constructing phenotypic maps for personalized medicine. This perspective argues that such dynamic predictive maps could also catalyze the development of personalized cancer treatment. They hold the potential to transform our understanding of cancer biology, providing a foundation for a future where therapy is tailored to each patient's unique molecular and cellular tumor profile. As our knowledge of cancer expands, these maps can be continually refined, ensuring they remain a valuable tool in precision oncology.
Collapse
Affiliation(s)
- Sayantan Bhattacharyya
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shafqat F. Ehsan
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Loukia G. Karacosta
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
31
|
Fuchs B, Studer G, Bode-Lesniewska B, Heesen P. The Next Frontier in Sarcoma Care: Digital Health, AI, and the Quest for Precision Medicine. J Pers Med 2023; 13:1530. [PMID: 38003845 PMCID: PMC10672685 DOI: 10.3390/jpm13111530] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
The landscape of sarcoma care is on the cusp of a transformative era, spurred by the convergence of digital health and artificial intelligence (AI). This perspectives article explores the multifaceted opportunities and challenges in leveraging these technologies for value-based, precision sarcoma care. We delineate the current state-of-the-art methodologies and technologies in sarcoma care and outline their practical implications for healthcare providers, administrators, and policymakers. The article also addresses the limitations of AI and digital health platforms, emphasizing the need for high-quality data and ethical considerations. We delineate the promise held by the synergy of digital health platforms and AI algorithms in enhancing data-driven decision-making, outcome analytics, and personalized treatment planning. The concept of a sarcoma digital twin serves as an illustrative paradigm for this integration, offering a comprehensive, patient-centric view of the healthcare journey. The paper concludes with proposals for future research aimed at advancing the field, including the need for randomized controlled trials or target trial emulations and studies focusing on ethical and economic aspects. While the road to this transformative care is laden with ethical, regulatory, and practical challenges, we believe that the potential benefits far outweigh the obstacles. We conclude with a call to action for multidisciplinary collaboration and systemic adoption of these technologies, underscoring the urgency to act now for the future betterment of sarcoma care and healthcare at large.
Collapse
Affiliation(s)
- Bruno Fuchs
- Sarcoma Service, University Teaching Hospital LUKS, University of Lucerne, 6000 Lucerne, Switzerland
- Sarcoma Service, Kantonsspital Winterthur, 8400 Winterthur, Switzerland
| | - Gabriela Studer
- Sarcoma Service, University Teaching Hospital LUKS, University of Lucerne, 6000 Lucerne, Switzerland
| | - Beata Bode-Lesniewska
- Patho Enge, SSN Reference Sarcoma Pathology, University of Zurich, 8000 Zurich, Switzerland
| | - Philip Heesen
- University Hospital USZ, University of Zurich, 8000 Zurich, Switzerland
| | | |
Collapse
|
32
|
Moerdler B, Krasner M, Orenbuch E, Grad A, Friedman B, Graber E, Barbiro-Michaely E, Gerber D. PTOLEMI: Personalized Cancer Treatment through Machine Learning-Enabled Image Analysis of Microfluidic Assays. Diagnostics (Basel) 2023; 13:3075. [PMID: 37835818 PMCID: PMC10572730 DOI: 10.3390/diagnostics13193075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Contemporary personalized cancer diagnostic approaches encounter multiple challenges. The presence of cellular and molecular heterogeneity in patient samples introduces complexities to analysis protocols. Conventional analyses are manual, reliant on expert personnel, time-intensive, and financially burdensome. The copious data amassed for subsequent analysis strains the system, obstructing real-time diagnostics at the "point of care" and impeding prompt intervention. This study introduces PTOLEMI: Python-based Tensor Oncological Locator Examining Microfluidic Instruments. PTOLEMI stands out as a specialized system designed for high-throughput image analysis, particularly in the realm of microfluidic assays. Utilizing a blend of machine learning algorithms, PTOLEMI can process large datasets rapidly and with high accuracy, making it feasible for point-of-care diagnostics. Furthermore, its advanced analytics capabilities facilitate a more granular understanding of cellular dynamics, thereby allowing for more targeted and effective treatment options. Leveraging cutting-edge AI algorithms, PTOLEMI rapidly and accurately discriminates between cell viability and distinct cell types within biopsy samples. The diagnostic process becomes automated, swift, precise, and resource-efficient, rendering it well-suited for point-of-care requisites. By employing PTOLEMI alongside a microfluidic cell culture chip, physicians can attain personalized diagnostic and therapeutic insights. This paper elucidates the evolution of PTOLEMI and showcases its prowess in analyzing cancer patient samples within a microfluidic apparatus. While the integration of machine learning tools into biomedical domains is undoubtedly in progress, this study's innovation lies in the fusion of PTOLEMI with a microfluidic platform-an integrated, rapid, and independent framework for personalized drug screening-based clinical decision-making.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Doron Gerber
- Life Sciences Faculty and Nanotechnology Institute, Bar-Ilan University, Ramat Gan 5290002, Israel
| |
Collapse
|
33
|
Mambetsariev I, Fricke J, Gruber SB, Tan T, Babikian R, Kim P, Vishnubhotla P, Chen J, Kulkarni P, Salgia R. Clinical Network Systems Biology: Traversing the Cancer Multiverse. J Clin Med 2023; 12:4535. [PMID: 37445570 PMCID: PMC10342467 DOI: 10.3390/jcm12134535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
In recent decades, cancer biology and medicine have ushered in a new age of precision medicine through high-throughput approaches that led to the development of novel targeted therapies and immunotherapies for different cancers. The availability of multifaceted high-throughput omics data has revealed that cancer, beyond its genomic heterogeneity, is a complex system of microenvironments, sub-clonal tumor populations, and a variety of other cell types that impinge on the genetic and non-genetic mechanisms underlying the disease. Thus, a systems approach to cancer biology has become instrumental in identifying the key components of tumor initiation, progression, and the eventual emergence of drug resistance. Through the union of clinical medicine and basic sciences, there has been a revolution in the development and approval of cancer therapeutic drug options including tyrosine kinase inhibitors, antibody-drug conjugates, and immunotherapy. This 'Team Medicine' approach within the cancer systems biology framework can be further improved upon through the development of high-throughput clinical trial models that utilize machine learning models, rapid sample processing to grow patient tumor cell cultures, test multiple therapeutic options and assign appropriate therapy to individual patients quickly and efficiently. The integration of systems biology into the clinical network would allow for rapid advances in personalized medicine that are often hindered by a lack of drug development and drug testing.
Collapse
Affiliation(s)
- Isa Mambetsariev
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Stephen B. Gruber
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Tingting Tan
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Razmig Babikian
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Pauline Kim
- Department of Pharmacy, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Priya Vishnubhotla
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Medical Oncology, City of Hope Atlanta, Newnan, GA 30265, USA
| | - Jianjun Chen
- Department of Systems Biology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Systems Biology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
34
|
Zhao J, Yabroff KR. High out‑of‑pocket spending and financial hardship at the end of life among cancer survivors and their families. Isr J Health Policy Res 2023; 12:24. [PMID: 37415261 DOI: 10.1186/s13584-023-00572-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 07/08/2023] Open
Abstract
Cancer is one of the most expensive medical conditions to treat worldwide, affecting national and local spending, as well as household budgets for patients and their families. In this commentary about a recent paper from Tur‑Sinai et al., we discuss the high out-of-pocket spending and medical and non-medical financial hardship faced by cancer patients and their families at the end-of-life in Israel. We provide recent information about the costs of health care in Israel and other high-income countries with (i.e., Canada, Australia, Japan, and Italy) and without universal health insurance coverage (i.e., United States, a country with high healthcare costs and uninsurance rate), and highlight the role of improving health insurance coverage and benefit design in reducing financial hardship among cancer patients and their families. Recognizing that financial hardship at the end of life affects both patients and their families, developing comprehensive programs and policies in Israel as well as in other countries is warranted.
Collapse
Affiliation(s)
- Jingxuan Zhao
- Surveillance and Health Equity Science, American Cancer Society, 3380 Chastain Meadows Pkwy NW Suite 200, Kennesaw, GA, 30144, USA.
| | - K Robin Yabroff
- Surveillance and Health Equity Science, American Cancer Society, 3380 Chastain Meadows Pkwy NW Suite 200, Kennesaw, GA, 30144, USA
| |
Collapse
|
35
|
From Pathogens to Cancer: Are Cancer Cells Evolved Mitochondrial Super Cells? Diagnostics (Basel) 2023; 13:diagnostics13040813. [PMID: 36832301 PMCID: PMC9954806 DOI: 10.3390/diagnostics13040813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/07/2023] [Accepted: 02/19/2023] [Indexed: 02/23/2023] Open
Abstract
Life is based on a highly specific combination of atoms, metabolism, and genetics which eventually reflects the chemistry of the Universe which is composed of hydrogen, oxygen, nitrogen, sulfur, phosphorus, and carbon. The interaction of atomic, metabolic, and genetic cycles results in the organization and de-organization of chemical information of that which we consider as living entities, including cancer cells. In order to approach the problem of the origin of cancer it is therefore reasonable to start from the assumption that the sub-molecular level, the atomic structure, should be the considered starting point on which metabolism, genetics, and external insults eventually emanate. Second, it is crucial to characterize which of the entities and parts composing human cells may live a separate life; certainly, this theoretical standpoint would consider mitochondria, an organelle of "bacteria" origin embedded in conditions favorable for the onset of both. This organelle has not only been tolerated by immunity but has also been placed as a central regulator of cell defense. Virus, bacteria, and mitochondria are also similar in the light of genetic and metabolic elements; they share not only equivalent DNA and RNA features but also many basic biological activities. Thus, it is important to finalize that once the cellular integrity has been constantly broken down, the mitochondria like any other virus or bacteria return to their original autonomy to simply survive. The Warburg's law that states the ability of cancers to ferment glucose in the presence of oxygen, indicates mitochondria respiration abnormalities may be the underlying cause of this transformation towards super cancer cells. Though genetic events play a key part in altering biochemical metabolism, inducing aerobic glycolysis, this is not enough to impair mitochondrial function since mitochondrial biogenesis and quality control are constantly upregulated in cancers. While some cancers have mutations in the nuclear-encoded mitochondrial tricarboxylic acid (TCA) cycle, enzymes that produce oncogenic metabolites, there is also a bio-physic pathway for pathogenic mitochondrial genome mutations. The atomic level of all biological activities can be considered the very beginning, marked by the electron abnormal behavior that consequently affects DNA of both cells and mitochondria. Whilst the cell's nucleus DNA after a certain number of errors and defection tends to gradually switch off, the mitochondria DNA starts adopting several escape strategies, switching-on a few important genes that belong back at their original roots as independent beings. The ability to adopt this survival trick, by becoming completely immune to current life-threatening events, is probably the beginning of a differentiation process towards a "super-power cell", the cancer cells that remind many pathogens, including virus, bacteria, and fungi. Thus, here, we present a hypothesis regarding those changes that first begin at the mitochondria atomic level to steadily involve molecular, tissue and organ levels in response to the virus or bacteria constant insults that drive a mitochondria itself to become an "immortal cancer cell". Improved insights into this interplay between these pathogens and mitochondria progression may disclose newly epistemological paradigms as well as innovative procedures in targeting cancer cell progressive invasion.
Collapse
|
36
|
Lopes-Júnior LC, Veronez LC. Personalized Care for Patients with Cancer in the Precision-Medicine Era. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3023. [PMID: 36833713 PMCID: PMC9957434 DOI: 10.3390/ijerph20043023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 05/25/2023]
Abstract
Important advances in cancer management have been made in the beginning of the 21st century [...].
Collapse
Affiliation(s)
- Luís Carlos Lopes-Júnior
- Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória 29043-900, ES, Brazil
| | - Luciana Chain Veronez
- Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto 14049-900, SP, Brazil
| |
Collapse
|
37
|
D'Errico S, Falanga AP, Greco F, Piccialli G, Oliviero G, Borbone N. State of art in the chemistry of nucleoside-based Pt(II) complexes. Bioorg Chem 2023; 131:106325. [PMID: 36577221 DOI: 10.1016/j.bioorg.2022.106325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/22/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
After the fortuitous discovery of the anticancer properties of cisplatin, many Pt(II) complexes have been synthesized, to obtain less toxic leads which could overcome the resistance phenomena. Given the importance of nucleosides and nucleotides as antimetabolites, studying their coordinating properties towards Pt(II) ions is challenging for bioorganic and medicinal chemistry. This review aims to describe the results achieved so far in the aforementioned field, paying particular attention to the synthetic aspects, the chemical-physical characterization, and the biological activities of the nucleoside-based Pt(II) complexes.
Collapse
Affiliation(s)
- Stefano D'Errico
- Department of Pharmacy, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy.
| | - Andrea Patrizia Falanga
- Department of Pharmacy, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy.
| | - Francesca Greco
- Department of Pharmacy, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy.
| | - Gennaro Piccialli
- Department of Pharmacy, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy.
| | - Giorgia Oliviero
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, via Sergio Pansini, 5, 80131 Naples, Italy.
| | - Nicola Borbone
- Department of Pharmacy, University of Naples Federico II, via Domenico Montesano, 49, 80131 Naples, Italy.
| |
Collapse
|
38
|
Parsonidis P, Beis G, Iliopoulos AC, Papasotiriou I. Adoptive transfer of activated immune cells against solid tumors: A preliminary study. Cell Immunol 2022; 382:104616. [PMID: 36219944 DOI: 10.1016/j.cellimm.2022.104616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND This study presents preliminary results concerning the effectiveness of a novel immunotherapy in cancer. The proposed adoptive cellular therapy product contains a mixture of effector immune cells, specifically macrophages, NK cells, dendritic cells, cytotoxic T lymphocytes and monoclonal antibody producing plasma cells. METHODS The results were based on both descriptive and inferential statistical analysis of data concerning 17 cancer patients. Particularly, performance scales such as clinical condition, Karnofsky-Index, ECOG index and symptom's scale were evaluated post therapy administration (4 months). Furthermore, circulating tumor cells (CTCs) and a specific tumor marker (EpCAM) were measured pre- and post-cellular therapy. RESULTS The results revealed a positive evaluation for clinical condition (70.59 %), Karnofsky-Index (88.23 %), ECOG index (94.12 %), and symptoms' scale (64.70 %). In addition, statistically significant reductions were found for both CTCs (p = 0.0016) and EpCAM positive cells (p = 0.0005), post-therapy, which were related to large size effects, namely 0.77 and 0.85, respectively. No cytokine storm, anaphylaxis or severe adverse events were observed with 4 months follow up evaluation. CONCLUSIONS These preliminary results indicate that the proposed cellular therapy can be considered for further studies in clinical trials.
Collapse
Affiliation(s)
| | - Georgios Beis
- Research Genetic Cancer Centre S.A., Florina, Greece
| | | | | |
Collapse
|
39
|
Fabro F, Kannegieter NM, de Graaf EL, Queiroz K, Lamfers MLM, Ressa A, Leenstra S. Novel kinome profiling technology reveals drug treatment is patient and 2D/3D model dependent in glioblastoma. Front Oncol 2022; 12:1012236. [PMID: 36408180 PMCID: PMC9670801 DOI: 10.3389/fonc.2022.1012236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma is the deadliest brain cancer. One of the main reasons for poor outcome resides in therapy resistance, which adds additional challenges in finding an effective treatment. Small protein kinase inhibitors are molecules that have become widely studied for cancer treatments, including glioblastoma. However, none of these drugs have demonstrated a therapeutic activity or brought more benefit compared to the current standard procedure in clinical trials. Hence, understanding the reasons of the limited efficacy and drug resistance is valuable to develop more effective strategies toward the future. To gain novel insights into the method of action and drug resistance in glioblastoma, we established in parallel two patient-derived glioblastoma 2D and 3D organotypic multicellular spheroids models, and exposed them to a prolonged treatment of three weeks with temozolomide or either the two small protein kinase inhibitors enzastaurin and imatinib. We coupled the phenotypic evidence of cytotoxicity, proliferation, and migration to a novel kinase activity profiling platform (QuantaKinome™) that measured the activities of the intracellular network of kinases affected by the drug treatments. The results revealed a heterogeneous inter-patient phenotypic and molecular response to the different drugs. In general, small differences in kinase activation were observed, suggesting an intrinsic low influence of the drugs to the fundamental cellular processes like proliferation and migration. The pathway analysis indicated that many of the endogenously detected kinases were associated with the ErbB signaling pathway. We showed the intertumoral variability in drug responses, both in terms of efficacy and resistance, indicating the importance of pursuing a more personalized approach. In addition, we observed the influence derived from the application of 2D or 3D models in in vitro studies of kinases involved in the ErbB signaling pathway. We identified in one 3D sample a new resistance mechanism derived from imatinib treatment that results in a more invasive behavior. The present study applied a new approach to detect unique and specific drug effects associated with pathways in in vitro screening of compounds, to foster future drug development strategies for clinical research in glioblastoma.
Collapse
Affiliation(s)
- Federica Fabro
- Department of Neurosurgery, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
| | | | | | | | - Martine L. M. Lamfers
- Department of Neurosurgery, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
| | | | - Sieger Leenstra
- Department of Neurosurgery, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, Netherlands
- *Correspondence: Sieger Leenstra,
| |
Collapse
|
40
|
Zhao J, Han X, Nogueira L, Fedewa SA, Jemal A, Halpern MT, Yabroff KR. Health insurance status and cancer stage at diagnosis and survival in the United States. CA Cancer J Clin 2022; 72:542-560. [PMID: 35829644 DOI: 10.3322/caac.21732] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 12/30/2022] Open
Abstract
Previous studies using data from the early 2000s demonstrated that patients who were uninsured were more likely to present with late-stage disease and had worse short-term survival after cancer diagnosis in the United States. In this report, the authors provide comprehensive data on the associations of health insurance coverage type with stage at diagnosis and long-term survival in individuals aged 18-64 years who were diagnosed between 2010 and 2013 with 19 common cancers from the National Cancer Database, with survival follow-up through December 31, 2019. Compared with privately insured patients, Medicaid-insured and uninsured patients were significantly more likely to be diagnosed with late-stage (III/IV) cancer for all stageable cancers combined and separately. For all stageable cancers combined and for six cancer sites-prostate, colorectal, non-Hodgkin lymphoma, oral cavity, liver, and esophagus-uninsured patients with Stage I disease had worse survival than privately insured patients with Stage II disease. Patients without private insurance coverage had worse short-term and long-term survival at each stage for all cancers combined; patients who were uninsured had worse stage-specific survival for 12 of 17 stageable cancers and had worse survival for leukemia and brain tumors. Expanding access to comprehensive health insurance coverage is crucial for improving access to cancer care and outcomes, including stage at diagnosis and survival.
Collapse
Affiliation(s)
- Jingxuan Zhao
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia
| | - Xuesong Han
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia
| | - Leticia Nogueira
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia
| | - Stacey A Fedewa
- Department of Hematology and Oncology, Emory University, Atlanta, Georgia
| | - Ahmedin Jemal
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia
| | - Michael T Halpern
- National Cancer Institute at the National Institutes of Health, Bethesda, Maryland
| | - K Robin Yabroff
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia
| |
Collapse
|
41
|
Alalawi M, Bakr AS, Reda R, Sadak KT, Nagy M. Late-onset toxicities of monoclonal antibodies in cancer patients. Immunotherapy 2022; 14:1067-1083. [PMID: 35892252 DOI: 10.2217/imt-2022-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cancer therapy duration is variable and may take years, adding a new challenge of maintaining the best life quality for cancer survivors. In cancer patients, late-onset toxicities have been reported with monoclonal antibodies and may involve several body organs or systems. They are defined as an autoimmune illnesses that can happen months to years after treatment discontinuation. Late-onset toxicities have become a focus of clinical care and related research. After cancer therapy is completed, the patient should receive longitudinal follow-up to detect these late effects as early as possible. The current review summarizes the recently reported late-onset toxicities of four classes of monoclonal antibodies (anti-CD52, anti-CTLA-4, anti-PD-1 and anti-CD20) with guidance for the diagnostic tools, appropriate management and treatment.
Collapse
Affiliation(s)
- Mai Alalawi
- Department of Pharmaceutical Services, Children's Cancer Hospital Egypt, Cairo, 57357, 4260102, Egypt.,Department of Pharmaceutical Sciences, Fakeeh College for Medical Sciences, Jeddah, 23323, Saudi Arabia
| | - Abrar Saeed Bakr
- Department of Pharmaceutical Services, Children's Cancer Hospital Egypt, Cairo, 57357, 4260102, Egypt.,Department of Clinical Pharmacy, Alexandria Vascular Center, Alexandria, 5431118, Egypt
| | - Rowaida Reda
- Department of Pharmaceutical Services, Children's Cancer Hospital Egypt, Cairo, 57357, 4260102, Egypt.,Department of Clinical Pharmacy, Woman Health Hospital, Assiut University, Assiut, 2074020, Egypt
| | - Karim Thomas Sadak
- University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA.,University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA
| | - Mohamad Nagy
- Department of Pharmaceutical Services, Children's Cancer Hospital Egypt, Cairo, 57357, 4260102, Egypt.,Personalized Medication Management Unit, Children's Cancer Hospital Egypt, Cairo, 57357, 4260102, Egypt
| |
Collapse
|
42
|
Jian H, Zhang Y, Wang J, Chen Z, Wen T. Zeolitic imidazolate framework-based nanoparticles for the cascade enhancement of cancer chemodynamic therapy by targeting glutamine metabolism. NANOSCALE 2022; 14:8727-8743. [PMID: 35674088 DOI: 10.1039/d2nr01736a] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The reprogrammed amino acid metabolism maintains the powerful antioxidant defense and DNA damage repair capacity of cancer cells, which could promote their escape from reactive oxygen species (ROS)-induced damage and inevitably diminish the efficacy of ROS-based therapies. Herein, we propose a strategy to enhance the effect of chemodynamic therapy (CDT) via glutaminolysis-targeted inhibition for cancer cells dependent on abnormal glutamine metabolism. To screen optimum drugs targeting glutamine metabolism, transcriptomic analysis is performed to identify predictive biomarkers. Eventually, telaglenastat (CB-839) is used to block mitochondrial glutaminase 1 (GLS 1) in basal-like breast cancer and loaded into the developed iron-doped zeolitic imidazolate frameworks (ZIF(Fe) NPs) to form ZIF(Fe)&CB nanoparticles, which are able to co-deliver Fe2+ and CB-839 into the tumor. CB-839 induced-glutaminolysis inhibition not only reduces intracellular antioxidants (glutathione, taurine) to amplify Fe2+-induced oxidative stress, but also decreases nucleotide pools (e.g., adenosine, dihydroorotate) to incur the deficiency of building blocks for DNA damage repair, thereby promoting the cell-killing effect of CDT. In vivo assessments further confirm the enhanced anticancer performance and good biocompatibility of ZIF(Fe)&CB nanoparticles. This study provides a promising strategy for the development and improvement of ROS-based anticancer nanosystems.
Collapse
Affiliation(s)
- Hui Jian
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yun Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing 100190, China
| | - Junyue Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenxiang Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Tingyi Wen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Innovation Academy for Green Manufacture, Chinese Academy of Sciences, Beijing 100190, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|