1
|
Fang Y, Hu F, Ren W, Xiang L, Wang T, Zhu C, He R, Dong X, Liu C, Ding H, Zhang K. Nanomedicine-unlocked radiofrequency dynamic therapy dampens incomplete radiofrequency ablation-arised immunosuppression to suppress cancer relapse. Biomaterials 2025; 317:123087. [PMID: 39778271 DOI: 10.1016/j.biomaterials.2025.123087] [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: 09/19/2024] [Revised: 12/16/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Incomplete radiofrequency ablation (iRFA) not only leaves residual tumor, but also render the residual tumor highly self-adaptable and immunosuppressive, consequently expediting residual tumor progression including relapse. To address it, radiofrequency dynamic therapy (RFDT) with identical trigger (namely radiofrequency) has been established and enabled by polyethylene glycol (PEG)-modified Fe-based single atom nanozyme (P@Fe SAZ). P@Fe SAZ can respond to radiofrequency field to produce reactive oxygen species (ROS), attaining the nanomedicine-unlocked low-temperature RFDT. Systematic experiments reveal that ROS further remodels iRFA-potentiated immunosuppressive microenvironment, e.g., expediting tumor-associated macrophages (TAMs) polarization into TAMs-M1, rejecting the intratumoral infiltrations of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). Coincidently, they have been demonstrated to stimulate dendritic cells (DCs) maturation and encourage the proliferations and infiltrations of effector T cells, consequently boosting anti-tumor immune responses and attenuating iRFA-enhanced plasticity, treatment resistance and self-adaptation of residual hepatocellular carcinoma (HCC) after iRFA. Thanks to them, such a nanomedicine-unlocked low-temperature RFDT exerts powerful actions on residual HCC model after iRFA with rapid expansion inhibition, relapse repression, survival prolongation, apoptosis promotion, etc. This low-temperature RFDT opens a window to address the iRFA-enhanced immunosuppression.
Collapse
Affiliation(s)
- Yan Fang
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, 200040, China. No. 12 Urumqi Middle Road, Shanghai 200040, China; Department of Laboratory Medicine and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Feixiang Hu
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, No. 270 Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Weiwei Ren
- Department of Laboratory Medicine and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China; Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301, Yanchangzhong Road, Shanghai, 200072, China
| | - Lihua Xiang
- Department of Laboratory Medicine and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China; Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301, Yanchangzhong Road, Shanghai, 200072, China
| | - Taixia Wang
- Department of Laboratory Medicine and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Chunyan Zhu
- Department of Laboratory Medicine and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Ruiqing He
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301, Yanchangzhong Road, Shanghai, 200072, China
| | - Xiulin Dong
- Department of Laboratory Medicine and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China
| | - Chang Liu
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301, Yanchangzhong Road, Shanghai, 200072, China.
| | - Hong Ding
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, 200040, China. No. 12 Urumqi Middle Road, Shanghai 200040, China.
| | - Kun Zhang
- Department of Laboratory Medicine and Department of Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Second Section, First Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
2
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [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: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
3
|
Alenezi SK. CAR T cells in lung cancer: Targeting tumor-associated antigens to revolutionize immunotherapy. Pathol Res Pract 2025; 269:155947. [PMID: 40168775 DOI: 10.1016/j.prp.2025.155947] [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: 01/18/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Tumor-targeted T cells engineered for targeting and killing tumor cells have revolutionized cancer treatment, specifically in hematologic malignancies, through chimeric antigen receptor (CAR) T cell therapy. However, the migration of this success to lung cancer is challenging due to the tumor microenvironment (TME), antigen heterogeneity, and limitations of T cell infiltration. This review aims to evaluate current strategies addressing these barriers, focusing on the optimization of tumor-associated antigen (TAA) targeting, such as epidermal growth factor receptor (EGFR), mucin-1 (MUC1), and mesothelin (MSLN), which are frequently overexpressed in lung cancer and offer promising targets for CAR T-cell therapy. In this review, we discuss recent progress in CAR T cell engineering, applying enhanced costimulatory molecules, cytokine-secreting CAR T cells, and engineered modifications to improve T cell resilience in immunosuppressive environments. Additionally, this review also evaluates combination therapies of immune checkpoint inhibitors and recently published clinical trials on lung cancer with CAR T cells. We offer insights into the way to optimize CAR T cell therapy for lung cancer by analyzing antigen selection, immune evasion, and the strategies to enhance T cell persistence and tumor infiltration.
Collapse
Affiliation(s)
- Sattam Khulaif Alenezi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia.
| |
Collapse
|
4
|
Lei H, Cui H, Xia Y, Sun F, Zhang W. Illuminating Hope for Tumors: The Progress of Light-Activated Nanomaterials in Skin Cancer. Int J Nanomedicine 2025; 20:5081-5118. [PMID: 40264819 PMCID: PMC12013650 DOI: 10.2147/ijn.s506000] [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: 11/12/2024] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
Skin cancer is a common malignant tumor that poses significant global health and economic burdens. The main clinical types include malignant melanoma and non-melanoma. Complications such as post-surgical recurrence, wound formation, or disfigurement can severely impact the patient's mental well-being. Traditional treatments such as surgery, chemotherapy, radiation therapy, and immunotherapy often face limitations. These challenges not only reduce the effectiveness of treatments but also negatively impact patients' quality of life. Phototherapy, a widely used and long-standing method in dermatology, presents a promising alternative for skin cancer treatment. Light-triggered nanomaterials further enhance the potential of phototherapy by offering advantages such as improved therapeutic precision, controlled drug release, minimal invasiveness, and reduced damage to surrounding healthy tissues. This review summarizes the application of light-triggered nanomaterials in skin cancer treatment, focusing on the principles, advantages, and design strategies of photodynamic therapy (PDT), photothermal therapy (PTT), and photoacoustic therapy (PAT). In this manuscript we have an in-depth discussion on overcoming translational barriers, including strategies to enhance light penetration, mitigate toxicity, reduce production costs, and optimize delivery systems. Additionally, we discuss the challenges associated with their clinical translation, including limited light penetration in deep tissues, potential toxicity, high production costs, and the need for advanced delivery systems.
Collapse
Affiliation(s)
- Huaqing Lei
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, People’s Republic of China
| | - Hengqing Cui
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Institute of Aesthetic Plastic Surgery and Medicine, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yu Xia
- College of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Fujia Sun
- College of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Wenjun Zhang
- Department of Burns and Plastic Surgery, Shanghai Changzheng Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
5
|
Zhong H, Li Y, Raza F, Xie J, Rong R, Qiu M, Su J. RBCs as a Bioinspired Drug Delivery System for Co-delivery of Irinotecan and Nanoalumina Enhances Colorectal Cancer Therapy. Mol Pharm 2025. [PMID: 40251121 DOI: 10.1021/acs.molpharmaceut.4c01396] [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: 04/20/2025]
Abstract
Colorectal cancer (CRC) is a malignant epithelial tumor with high morbidity and mortality. In CRC treatment, irinotecan (CPT-11) as a chemotherapeutic drug is widely applied. However, its half-life is short, leading to large dosages and severe side effects. Red blood cells (RBCs) are biocompatible drug carriers with high capacity, avoiding premature drug degradation and achieving slow drug release. Nanoalumina (AN) is an emerging immune adjuvant that can enhance the immune response. Here, we used RBCs as carriers and absorbed AN to construct AN-CPT-11-RBCs. CPT-11 would induce tumor cell death, releasing much tumor antigen, while AN would activate immune cells to recognize newly released antigens and induce lymphocyte proliferation, enhancing the antitumor effect simultaneously. With loading amounts of 4 mg of CPT-11 and 3 mg of AN per 109 RBCs, AN-CPT-11-RBCs had similar properties to natural RBCs. In vivo, AN-CPT-11-RBCs could circulate for 9 days and stimulate the proliferation of lymphocytes in the spleen and tumor tissue, having a higher tumor growth inhibition rate of 74.01% and a lower frequency of administration. In conclusion, AN-CPT-11-RBCs attain the co-delivery of CPT-11 and AN for the synergistic treatment of CRC.
Collapse
Affiliation(s)
- Hongyu Zhong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yichen Li
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiyuan Xie
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruonan Rong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
6
|
Hemida N, El-Gamil DS, ElHady AK, Lin KC, Chang YH, Hilscher S, Schutkowski M, Ibrahim HS, Hamed MM, Chen SH, Chen CH, Abadi AH, Sippl W, Chen PJ, Cheng YS, Abdel-Halim M. Unlocking the potential of novel tetrahydro-β-carboline-based HDAC6 inhibitors for colorectal cancer therapy: Design, synthesis and biological evaluation. Bioorg Chem 2025; 160:108454. [PMID: 40252366 DOI: 10.1016/j.bioorg.2025.108454] [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: 01/25/2025] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025]
Abstract
Altered histone deacetylase 6 (HDAC6) expression and function have been linked to cancer progression, positioning it as a promising therapeutic target for cancer treatment. Herein, we introduce HDAC6 inhibitors based on the tetrahydro-β-carboline scaffold, with compound 18d exhibiting the strongest HDAC6 inhibitory potency, achieving an IC50 of 1.3 nM. Compound 18d exhibited significant growth inhibitory activity against an NCI panel of 60 human cancer cell lines with a minimal cytotoxic effect on non-tumor cells. In vitro mechanistic investigations were conducted in HCT-116 colorectal cancer cells where the capability of 18d to enhance the acetylation of α-tubulin (HDAC6 substrate) rather than nuclear H3 histone (HDAC1 substrate) confirmed selective inhibition of HDAC6 subtype. Additionally, compound 18d was observed to suppress the S phase and promote accumulation in the apoptotic sub-G1 phase, potentially through increasing cleaved caspase 3 and reducing Bcl-2 levels in HCT-116 cells. A wound healing assay also elicited the ability of 18d to hinder cell migration. Notably, 18d could suppress the phosphorylation of extracellular signal-regulated kinase (ERK)1/2, a crucial signaling pathway implicated in cancer cell proliferation, migration and apoptosis. Moreover, downregulation of the critical immune checkpoint protein programmed death-ligand 1 (PD-L1) revealed a potential role of 18d in augmenting immune response towards tumor cells. In summary, these findings highlight 18d's dual role in direct tumor growth suppression and immune system sensitization, highlighting a broader cancer therapeutic potential beyond conventional HDAC inhibition.
Collapse
Affiliation(s)
- Noreen Hemida
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Dalia S El-Gamil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; Department of Chemistry, Faculty of Pharmacy, Ahram Canadian University, Cairo 12451, Egypt
| | - Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt; School of Life & Medical Sciences, University of Hertfordshire hosted by Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Kai-Chun Lin
- Institute of Plant Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Yen-Hua Chang
- Institute of Plant Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Sebastian Hilscher
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Mike Schutkowski
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Hany S Ibrahim
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Mostafa M Hamed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany
| | - Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831301, Taiwan
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital and Graduate Institute of Medicine, I-Shou University, Kaohsiung 824410, Taiwan
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg, Halle (Saale), Germany
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital and Graduate Institute of Medicine, I-Shou University, Kaohsiung 824410, Taiwan; Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Yi-Sheng Cheng
- Institute of Plant Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan; Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt.
| |
Collapse
|
7
|
Ma S, Lu Y, Sui S, Yang JS, Fu BB, Tan PX, Chai Y, Lv J, Kong L, Wu X, Gao YB, Yan T. Unraveling the triad of immunotherapy, tumor microenvironment, and skeletal muscle biomechanics in oncology. Front Immunol 2025; 16:1572821. [PMID: 40242775 PMCID: PMC12000078 DOI: 10.3389/fimmu.2025.1572821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/18/2025] Open
Abstract
The intricate interaction between skeletal muscle biomechanics, the tumor microenvironment, and immunotherapy constitutes a pivotal research focus oncology. This work provides a comprehensive review of methodologies for evaluating skeletal muscle biomechanics, including handheld dynamometry, advanced imaging techniques, electrical impedance myography, elastography, and single-fiber experiments to assess muscle quality and performance. Furthermore, it elucidates the mechanisms, applications, and limitations of various immunotherapy modalities, including immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and combined chemoimmunotherapy, while examining their effects on skeletal muscle function and systemic immune responses. Key findings indicate that although immunotherapy is effective in augmenting antitumor immunity, it frequently induces muscle-related adverse effects such as weakness, fatigue, or damage, primarily mediated by cytokine release and immune activation. This work underscores the significance of immune niches within the tumor microenvironment in influencing treatment outcomes and proposes strategies to optimize therapy through personalized regimens and combinatorial approaches. This review highlights the need for further research on the formation of immune niches and interactions muscle-tumor. Our work is crucial for advancing the efficacy of immunotherapy, reducing adverse effects, and ultimately improving survival rates and quality of life of patients with cancer.
Collapse
Affiliation(s)
- Shuang Ma
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Ying Lu
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Shang Sui
- St. John’s Kilmarnock School, Breslau, ON, Canada
| | - Jia-shuo Yang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing-bing Fu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-xin Tan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Chai
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Jiaqi Lv
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Lingyu Kong
- School of Information Science and Engineering, Shenyang Ligong University, Shenyang, China
| | - Xiaolin Wu
- School of Mathematics and Statistics, Liaoning University, Shenyang, China
| | - Yi-bo Gao
- Department of Oral and Maxillofacial Surgery, Taikang Bybo Dental, Beijing, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Sareen G, Mohan M, Mannan A, Dua K, Singh TG. A new era of cancer immunotherapy: vaccines and miRNAs. Cancer Immunol Immunother 2025; 74:163. [PMID: 40167762 PMCID: PMC11961864 DOI: 10.1007/s00262-025-04011-5] [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: 01/23/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has transformed the treatment landscape, introducing new strategies to fight various types of cancer. This review examines the important role of vaccines in cancer therapy, focusing on recent advancements such as dendritic cell vaccines, mRNA vaccines, and viral vector-based approaches. The relationship between cancer and the immune system highlights the importance of vaccines as therapeutic tools. The discussion covers tumor cell and dendritic cell vaccines, protein/peptide vaccines, and nucleic acid vaccines (including DNA, RNA, or viral vector-based), with a focus on their effectiveness and underlying mechanisms. Combination therapies that pair vaccines with immune checkpoint inhibitors, TIL therapy, and TCR/CAR-T cell therapy show promising potential, boosting antitumor responses. Additionally, the review explores the regulatory functions of microRNAs (miRNAs) in cancer development and suppression, featuring miR-21, miR-155, the let-7 family, and the miR-200 family, among others. These miRNAs influence various pathways, such as PI3K/AKT, NF-κB, and EMT regulation, providing insights into biomarker-driven therapeutic strategies. Overall, this work offers a thorough overview of vaccines in oncology and the integrative role of miRNAs, setting the stage for the next generation of cancer immunotherapies.
Collapse
Affiliation(s)
- Gitika Sareen
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
9
|
Khan M, Ullah R, Wang G, Chu M. Macrophage membrane-functionalized nanotherapeutics for tumor targeted therapy. Theranostics 2025; 15:4823-4847. [PMID: 40225567 PMCID: PMC11984399 DOI: 10.7150/thno.108875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is a multifaceted disease characterized by uncontrollable cell growth. To date, various therapies are employed including conventional chemotherapy, surgery, radiotherapy, and immunotherapies. However, these approaches still present significant limitations. Interestingly, macrophage membranes utilize their innate antigen recognition affinity to facilitate targeted localization to tumor sites with high specificity. As a result, they display distinct characteristics such as avoiding premature leakage, tumor targeting ability, immune evasion, immune system activation, tumor-infiltrating ability, improved cell endocytosis and release payload in tumor-microenvironment. In this paper, the recent advances in macrophage-membrane-encapsulated nanotherapeutics for targeted cancer therapy are presented. We begin by introducing macrophage membrane-encapsulated nanotherapeutics preparation and characterization, followed by cancer immunotherapy such as macrophage polarization, T-cell infiltration, macrophage membrane modification, immunization, and inducing immunological cell death. Lastly, a future perspective is proposed to highlight the limitations of macrophage membrane-encapsulated nanotherapeutics and the possible resolutions toward the clinical transformation of currently developed biomimetic chemotherapies. We believe this review may be beneficial for improving the deep research of macrophage membrane-encapsulated nanotherapeutics for targeted cancer therapy.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, P.R. China
| | - Razi Ullah
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Jinfeng Laboratory, Chongqing, 400030, P.R. China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Jinfeng Laboratory, Chongqing, 400030, P.R. China
| | - Maoquan Chu
- Research Center for Translational Medicine at Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P.R. China
| |
Collapse
|
10
|
Yang Y, Wu Y. Potential of bacterial outer membrane vesicles in tumor vaccine: characteristics, advancements, and future directions. Essays Biochem 2025; 69:EBC20253004. [PMID: 40159726 DOI: 10.1042/ebc20253004] [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: 01/03/2025] [Accepted: 03/02/2025] [Indexed: 04/02/2025]
Abstract
Bacterial outer membrane vesicles (OMVs), naturally released by Gram-negative bacteria, are a type of lipid bilayer nanoparticles containing many components found within the parent bacterium. Despite OMVs were first considered mere by-products of bacterial growth, recent studies have shown them as a highly adaptable platform for tumor vaccine. Here, we first demonstrate the biogenesis of OMVs, then review the strong immunogenicity of OMVs as an immune adjuvant in tumor vaccine and its excellent vaccine delivery capability, and finally discuss OMVs' engineering potentials through summarizing recent scientific advancements in genetic engineering, chemical modification, and nanotechnology. We also point out the clinical trials and future challenges of OMV-based vaccine. Overall, this review offers valuable insights into cancer immunotherapy, providing a roadmap for leveraging OMVs as a versatile platform for next-generation cancer vaccines.
Collapse
Affiliation(s)
- Yizhe Yang
- College of Nano Science & Technology (CNST), Soochow University, Suzhou, Jiangsu 215123, China
| | - Yumin Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
11
|
Dhasmana A, Santhanam A, Dhasmana K, Malik S, Preetam S. Innovative smart biosensors for cancer theranostics: A new frontier in detection, diagnosis, and beyond. Cancer Treat Res Commun 2025; 43:100911. [PMID: 40156955 DOI: 10.1016/j.ctarc.2025.100911] [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: 12/13/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Cancer is still a major health concern worldwide, requiring ongoing improvements in methods of diagnosis and treatment. During the past decade, smart biosensors have become essential instruments in cancer theranostics, improving diagnosis accuracy, tracking treatment efficacy, and customizing patient care. This review thoroughly investigates how smart biosensors have revolutionized the field of cancer. The potential of major technical advancements, such as wearable technology, microfluidic platforms, and sensors based on nanomaterials to identify cancer biomarkers with high sensitivity and specificity is investigated. A detailed discussion is held regarding clinical applications that include early diagnosis, real-time monitoring of therapy responses, and support for personalized medicine techniques. Future directions targeted at optimizing the therapeutic utility of smart biosensors in oncology are also examined, along with issues pertaining to regulatory routes and clinical translation hurdles. This study highlights the potential of smart biosensors to transform cancer treatment, bringing in a new era of precision medicine and better patient outcomes by combining insights from multiple viewpoints.
Collapse
Affiliation(s)
- Archna Dhasmana
- Himalayan School of Bioscience, Swami Rama Himalayan University, Jolly Grant, Dehradun Uttarakhand 248140, India.
| | - Ayushi Santhanam
- Himalayan School of Bioscience, Swami Rama Himalayan University, Jolly Grant, Dehradun Uttarakhand 248140, India
| | - Khushi Dhasmana
- Himalayan School of Bioscience, Swami Rama Himalayan University, Jolly Grant, Dehradun Uttarakhand 248140, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi 834001, India; School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand 248007, India; University Center for Research & Development (UCRD) Chandigarh University, NH-05 Chandigarh- Ludhiana Highway, Mohali, Punjab 140413, India.
| | - Subham Preetam
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika, 59053, Sweden.
| |
Collapse
|
12
|
Narote S, Desai SA, Patel VP, Deshmukh R, Raut N, Dapse S. Identification of new immune target and signaling for cancer immunotherapy. Cancer Genet 2025; 294-295:57-75. [PMID: 40154216 DOI: 10.1016/j.cancergen.2025.03.004] [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: 01/01/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy has become one of the innovative treatments in malignancy as it activates the immune system to find and eliminate malignant cells. The tumor immunology interface has become increasingly intricate, making the identification of new immune targets and signalling pathways on which to base improved therapeutic strategies an ongoing process. This review, we goal to clarify the contacts between cancer and immune system with a focus on immune surveillance as well as immune evasion mechanisms. Comprehensive immunotherapeutic therapies are overviewed with ICI (CTLA-4, PD-1, PD-L1), CAR-T cell therapy, and cancer vaccines whereas, advanced therapies targeting new immune checkpoints are also elucidated including TIM-3, LAG-3, and TIGIT. The JAK/STAT, MAPK and PI3K-AKT-mTOR pathways are reviewed with regards to cancer progression and immunotherapeutic resistance. The dysregulation of these pathways gives hope for the identification of fresh targets for therapy. Genomics, proteomics, immunopeptidomics, single cell mass spectrometry, CRISPR-based functional genomics and bioinformatics are described as essential for immune target identification and for mapping of cancer relevant signaling pathways. This review also considers some emerging issues in the subject area like the tumor heterogeneity, immune-related adverse events (irAEs), and personalized treatment. These barriers are described to facilitate the understanding of ways to overcome them and increase the efficacy of immunotherapies through combination therapies. This means that by developing new knowledge of immunological targets and pathways, immunoprecision medicine for cancer could greatly enhance outcomes.
Collapse
Affiliation(s)
- Sakshi Narote
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India.
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Rutuja Deshmukh
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nikita Raut
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sejal Dapse
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| |
Collapse
|
13
|
Liu H, Ding S, Lyu W, Lu S, Liu X. Chito-oligosaccharide impairs the proliferation, invasion and migration of pancreatic cancer cells. Discov Oncol 2025; 16:298. [PMID: 40069446 PMCID: PMC11896950 DOI: 10.1007/s12672-025-02015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Chito-oligosaccharide (COS) is a low molecular weight polymer obtained by degrading chitosan through special enzymatic technology, with good water solubility and high biological activity. It is also the only positively charged cationic basic amino oligosaccharide in nature. Studies have confirmed that COS has antitumour effect, but research on its effect on pancreatic cancer (PC) remains limited and unclear. This study aimed to explore the effects of COS on PC cells (PANC-1 and MIAPaCa-2). METHOD We used different concentrations of COS to treat PC cells and conducted Cell Counting Kit-8, wound-healing, and transwell assays to evaluate the proliferation, invasion, and migration ability of PC cells, respectively. Western blot was conducted to assess the expression levels of epithelial-mesenchymal transition (EMT) related markers. RESULT The proliferation, invasion, and migration ability of PC cells (PANC-1 and MIAPaCa-2) gradually decreased in a manner dependent on COS concentration. COS at 10 mg/mL exerted the strongest inhibitory effect on the two PC cell lines. At 10 mg/mL, the proliferative activity was 60.61% ± 5.25% and 64.02% ± 4.96%, respectively; the invasive ability was (18.67 ± 4.416) and (31.33 ± 3.162), respectively; and the cell-migration ability was 26.83% ± 0.442% and 17.66% ± 0.647%, respectively. The expression levels of N-cadherin and vimentin were significantly downregulated in PANC-1 cells (0.198 ± 0.047 and 0.225 ± 0.038, respectively) and MIAPaCa-2 cells (0.214 ± 0.094 and 0.214 ± 0.094, respectively) at 10 mg/mL, respectively. Conversely, E-cadherin was upregulated (0.460 ± 0.037 and 0.491 ± 0.047, respectively). Compared with control group, the differences were statistically significant. CONCLUSION The upregulation of E-cadherin and the downregulation of vimentin and N-cadherin suggested that the specific mechanism of COS in PC may be related to EMT. This study provided a new direction for PC treatment.
Collapse
Affiliation(s)
- Han Liu
- Department of Gastroenterology, General Hospital of Northern Theatre Command, 83 Wenhua Road, Shenyang, 110840, Liaoning, China
| | - Siyuan Ding
- Department of Gastroenterology, General Hospital of Northern Theatre Command, 83 Wenhua Road, Shenyang, 110840, Liaoning, China
| | - Weiyan Lyu
- Department of Gastroenterology, General Hospital of Northern Theatre Command, 83 Wenhua Road, Shenyang, 110840, Liaoning, China
| | - Shengyan Lu
- Department of Gastroenterology, General Hospital of Northern Theatre Command, 83 Wenhua Road, Shenyang, 110840, Liaoning, China
| | - Xu Liu
- Department of Gastroenterology, General Hospital of Northern Theatre Command, 83 Wenhua Road, Shenyang, 110840, Liaoning, China.
| |
Collapse
|
14
|
Zhang Z, Xu C, Gong N, Qing G, Zhang Y, Shi Y, Brenner JS, Li F, Xu FJ, Liang XJ. An antigen-capturing and lymph node-targeting nanoparticle for cancer immunotherapy. J Control Release 2025; 379:993-1005. [PMID: 39889883 DOI: 10.1016/j.jconrel.2025.01.087] [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: 10/14/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Cancer immunotherapy leverages the immune system to combat cancer and has shown promise for many patients. However, its effectiveness is often hampered by an immunosuppressive tumor microenvironment and the low immunogenicity of tumor cells. In this study, we developed an in situ cancer vaccine that integrates chemotherapy and immunotherapy in a single platform. We synthesized two amphiphilic polymers with poly-albumin-binding domains (PABD) that can target the lymph nodes, PABD-PGEA and PABD-PGED. Compared with previous albumin-hijacking strategies utilizing the same albumin-binding domains, PABD-PGEA exhibited approximately six times greater lymph node-targeting ability, demonstrating enhanced antigen-capturing capability. We loaded PABD-PGEA with doxorubicin (DOX), a drug known to induce immunogenic cell death (ICD) in tumor cells, to form DOX@PABD-PGEA nanomicelles. DOX@PABD-PGEA inhibited tumor growth and extended the survival of mice with B16F10 melanoma through chemotherapy and immunotherapy. Notably, DOX@PABD-PGEA prevented tumor recurrence post-surgery by promoting efficient antigen presentation and reversing immunosuppression in the tumor microenvironment. Our findings suggest that DOX@PABD-PGEA, as an antigen-capturing nanoparticle, provides a safe and effective platform for in situ cancer vaccines and improves cancer immunotherapy.
Collapse
Affiliation(s)
- Zhen Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Chen Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ningqiang Gong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Guangchao Qing
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Yuxuan Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Yuxuan Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fangzhou Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Fu-Jian Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
15
|
Köylü B, Esen BH, Bektaş ŞN, Özbek L, Turan V, Urman B, Öktem Ö, Selçukbiricik F. Pharmacovigilance analysis of immune checkpoint inhibitor-related reproductive adverse effects based on the FDA adverse event reporting system. Sci Rep 2025; 15:7770. [PMID: 40044844 PMCID: PMC11883018 DOI: 10.1038/s41598-025-91476-0] [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: 10/08/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025] Open
Abstract
This study aims to investigate the adverse effects of immune checkpoint inhibitors (ICIs) on the female and male reproductive systems. In the FDA Adverse Event Reporting System (FAERS) database, adverse reactions under the "Reproductive system and breast disorders" category in the System Organ Classes were included, covering a period from January 1, 2015, to June 30, 2023. We identified 133,512 patients treated with ICIs. Immune checkpoint inhibitor-related reproductive adverse effects (irRAEs) were reported in 568 (0.43%) patients. Spermatogenesis abnormality (ROR025 = 7.91) had the highest signal strength associated with ICI use in males. Genital tract fistula was the only significant irRAE (ROR025 = 2.72) in females. PD-1 inhibitors pose greater risk than CTLA-4 inhibitors (OR = 1.65 [1.05-2.79], p = 0.045). Gynecologic cancers in females (OR = 3.77 [2.82-4.99], p < 0.0001) and urogenital cancers in males (OR = 1.56 [1.17-2.06], p = 0.0018) carried the highest risk compared to other cancers. Additional targeted drugs (OR = 2.32 [1.76-3.02], p < 0.0001), particularly lenvatinib (OR = 3.50 [2.48-4.94], p < 0.0001) and cabozantinib (OR = 3.71 [1.96-7.03], p < 0.0001) significantly increased the risk for females. Additional use of chemotherapy drugs was associated with a significant reduction in the risk for males (OR = 0.65 [0.42-0.96], p = 0.042) except for doxorubicin (OR = 2.58 [1.22-5.47], p = 0.013) and cyclophosphamide (OR = 2.36 [1.05-5.29], p = 0.038). This study demonstrates that ICIs could potentially lead to a wide range of adverse effects in the reproductive system in both males and females.
Collapse
Affiliation(s)
- Bahadır Köylü
- Department of Internal Medicine, Division of Medical Oncology, Koç University School of Medicine, Istanbul, Turkey.
- Department of Internal Medicine, Division of Medical Oncology, Koç University Hospital, 34010, Topkapi, Istanbul, Turkey.
| | | | | | - Laşin Özbek
- Koç University School of Medicine, Istanbul, Turkey
| | - Volkan Turan
- Istanbul Health and Technology University Faculty of Medicine, Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Bülent Urman
- Department of Obstetrics and Gynecology, Koç University School of Medicine, Istanbul, Turkey
| | - Özgür Öktem
- Department of Obstetrics and Gynecology, Koç University School of Medicine, Istanbul, Turkey.
- Department of Obstetrics and Gynecology, Koç University Hospital, 34010, Topkapi, Istanbul, Turkey.
| | - Fatih Selçukbiricik
- Department of Internal Medicine, Division of Medical Oncology, Koç University School of Medicine, Istanbul, Turkey
| |
Collapse
|
16
|
Krass M, Kolster M, Valenzuela JI, Moldenhauer L, Kagelmacher M, Niesler N, Weng A, Zerial M, Nagel G, Fuchs H. Recombinant Expression of a Ready-to-Use EGF Variant Equipped With a Single Conjugation Site for Click-Chemistry. Eng Life Sci 2025; 25:e70015. [PMID: 40104837 PMCID: PMC11913717 DOI: 10.1002/elsc.70015] [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: 07/24/2024] [Revised: 01/02/2025] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
The epidermal growth factor (EGF) receptor is commonly targeted in cancer therapy because it is overexpressed in many malignant cells. However, a general problem is to couple the targeting moieties and the drug molecules in a way that results in a homogeneous product. Here, we overcome this issue by engineering a variant of EGF with a single conjugation site for coupling virtually any payload. The recombinant EGF variant K-EGFRR was expressed in E. coli Rosetta with a 4-6 mg/L yield. To confirm the accessibility of the introduced functional group, the ligand was equipped with a sulfo-cyanine dye with a loading of 0.65 dye per ligand, which enables tracking in vitro. The kinetics and affinity of ligand-receptor interaction were evaluated by enzyme-linked immunosorbent assay and surface plasmon resonance. The affinity of K-EGFRR was slightly higher when compared to the wild-type EGF (K D: 5.9 vs. 7.3 nM). Moreover, the ligand-receptor interaction and uptake in a cellular context were evaluated by flow cytometry and quantitative high-content imaging. Importantly, by attaching heterobifunctional polyethylene glycol linkers, we allowed orthogonal click-conjugation of the ligand to any payload of choice, making K-EGFRR an ideal candidate for targeted drug delivery.
Collapse
Affiliation(s)
- Melanie Krass
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin Berlin Germany
| | - Meike Kolster
- Institut für Pharmazie Freie Universität Berlin Berlin Germany
| | | | - Lena Moldenhauer
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin Berlin Germany
| | - Marten Kagelmacher
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin Berlin Germany
- Institut für Chemie und Biochemie Freie Universität Berlin Berlin Germany
| | - Nicole Niesler
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin Berlin Germany
| | - Alexander Weng
- Institut für Pharmazie Freie Universität Berlin Berlin Germany
| | - Marino Zerial
- Max Planck Institute of Molecular Cell Biology and Genetics Dresden Germany
| | - Gregor Nagel
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin Berlin Germany
| | - Hendrik Fuchs
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin Berlin Germany
| |
Collapse
|
17
|
Zhao Y, Cheng J, Li Z, Wang J, Chen X. Nanozymes in Biomedical Applications: Innovations Originated From Metal-Organic Frameworks. Adv Healthc Mater 2025; 14:e2402066. [PMID: 39319491 DOI: 10.1002/adhm.202402066] [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: 06/05/2024] [Revised: 08/18/2024] [Indexed: 09/26/2024]
Abstract
Nanozymes exhibit significant potential in medical theranostics, environmental protection, energy development, and biopharmaceuticals due to their exceptional catalytic performance. Compared with natural enzymes, nanozymes have the advantages of simple preparation and purification, convenient production and low cost. Therefore, it is very important to prepare nanozymes quickly and efficiently, which not only helps to expand their application scope, but also can further exert their great potential in various fields. Metal-organic frameworks (MOF) materials serve as versatile substrates for constructing nanozymes, offering unique advantages like adjustable structure, high specific surface area, and porous channels. MOF coordination nodes constructed from metal ions or metal clusters have unique properties that can be leveraged to tailor nanozyme characteristics for different applications. This review describes and analyzes recent methods for constructing nanozymes using MOF materials, and explores their application prospects in biomedicine. By expounding the preparation techniques and biomedical applications of nanozymes, this review aims to inspire researchers to develop innovative nanozyme materials and explore new application directions.
Collapse
Affiliation(s)
- Yuewu Zhao
- College of Pharmacy, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou, 253023, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Junjie Cheng
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Zhen Li
- College of Pharmacy, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou, 253023, China
| | - Jine Wang
- College of Pharmacy, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou, 253023, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| |
Collapse
|
18
|
Abohassan M, Shahrani MMA, Alouda SK, Rajagopalan P. Dibenzo [a, c] phenazin-11-yl(phenyl) methanone (SBLJ23), a novel selective inhibitor targeting JAK2 V617F mutation in myeloproliferative neoplasms. Oncol Res 2025; 33:675-685. [PMID: 40109858 PMCID: PMC11915075 DOI: 10.32604/or.2024.056256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 03/22/2025] Open
Abstract
Background The JAK2V617F mutation plays a crucial part in the pathogenesis of myeloproliferative neoplasms (MPN), which includes polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) leading to aberrant proliferation and survival of hematopoietic cells. Alongside the challenges of drug resistance and side effects, identifying novel compounds that selectively target JAK2V617F could provide more effective and safer therapeutic options for patients with MPNs. Materials and Methods We employed computational approaches like high-throughput virtual screening, molecular dynamics simulations (MDS), and binding free energy calculations to identify inhibitors targeting wild and mutant JAK2 kinases. JAK2V617F positive HEL, wild type JAK2 positive TF-1, and non-cancerous Vero cells were used for in vitro validations. Results SBLJ23 emerged as a top candidate inhibitor with specificity for JAK2V617F. Protein-ligand interaction studies and MDS revealed stable interactions and binding of SBLJ23 over the simulation period, with Root Mean Square Deviation (RMSD) indicating consistent binding after 1t15ns. SBLJ23 displayed a half maximal inhibitory concentration (IC50) value of 522.4 nM against the JAK2 enzyme. The compound exhibited inhibition of cell proliferation in HEL and TF-1 cells, with half maximal cell growth inhibitory concentration (GI50) values of 2.51 and 15.87 µM, respectively. Moreover, SBLJ23 induced G2/M cell cycle arrest in HEL cells to facilitate apoptosis in these cell lines. The compound significantly reduced the percentage of phospho JAK2 and phospho STAT3 in HEL cells. Conclusion High binding affinity, stable interaction profile, favorable binding free energy, and in vitro validations claim SBLJ23 as a potential lead compound against JAK2V617F and suggest further development and optimization towards clinical application in managing myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| | - Mesfer Mohammad Al Shahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| | - Sarah Khaled Alouda
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| | - Prasanna Rajagopalan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| |
Collapse
|
19
|
Chand T, Dubey AK, Misra G. Unraveling HPV-associated cancer complexity: From molecular insights to innovative therapies. Heliyon 2025; 11:e42437. [PMID: 40007779 PMCID: PMC11850150 DOI: 10.1016/j.heliyon.2025.e42437] [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: 12/19/2023] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Human papillomavirus (HPV) contributes to a high global incidence of sexually transmitted infections, predominantly associated with cervical cancer, as well as head and neck, penile, anal, vaginal, and vulvar cancers. Despite efforts through improved screening and HPV vaccination campaigns, challenges persist, influencing the frequency of HPV-related malignancies. Collaborative scientific endeavors strive to pioneer groundbreaking approaches, aiming to alleviate the adverse consequences of HPV-related malignancies on individuals and communities. The present review is focused on exhaustively covering HPV-associated cancers, particularly cervical cancer. This study highlights the initiation, progression, immune invasion, and treatment strategies of HPV-associated cancers. The role of viral oncoproteins E6 and E7 responsible for immune evasion and subsequent latent infection is also elaborated. The article also sheds light on the pivotal role of HPV vaccination in averting high-risk HPV infections and associated cancers. The scope of this review encompasses HPV-associated cancer epidemiology, regional disparities, and the distinctive challenges faced in the context of India. This will be a value addition to the knowledge repertoire beneficial for creating awareness and designing health policies.
Collapse
Affiliation(s)
- Tara Chand
- National Institute of Biologicals (Ministry of Health & Family Welfare, Government of India), A-32, Sector-62, Noida, 201309, India
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201 002, India
| | - Ashwini Kumar Dubey
- National Institute of Biologicals (Ministry of Health & Family Welfare, Government of India), A-32, Sector-62, Noida, 201309, India
| | - Gauri Misra
- National Institute of Biologicals (Ministry of Health & Family Welfare, Government of India), A-32, Sector-62, Noida, 201309, India
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, 201 002, India
| |
Collapse
|
20
|
Jo A, Shoji T, Otsuka H, Abe M, Tatsuki S, Chiba Y, Sato S, Takatori E, Kaido Y, Nagasawa T, Kagabu M, Baba T. Treatment strategies for advanced and recurrent endometrial cancer using immune checkpoint inhibitors. Int J Clin Oncol 2025; 30:229-240. [PMID: 39812928 DOI: 10.1007/s10147-024-02689-8] [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: 08/02/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Doxorubicin + cisplatin and paclitaxel + carboplatin are standard chemotherapy regimens for endometrial cancer. The development of PD-1 and PDL-1 antibody drugs has led to the use of these agents for endometrial cancer in other countries. The KEYNOTE-775 trial for advanced or recurrent endometrial cancer demonstrated the benefits of pembrolizumab and lenvatinib combination therapy, and the results of this trial led to the approval of its coverage for recurrent cancer by the Japanese health insurance system. Currently, treatment with immune checkpoint inhibitors is transitioning from second-line to first-line therapy. In a global randomized phase III study, the drugs dostarlimab, durvalumab, and atezolizumab, which are not yet approved in Japan, showed better results in the study arms than in the control arm. Additionally, biomarkers have been developed for endometrial cancer, enabling gynecologists to pursue treatment options based on the biomarkers detected for better treatment outcomes. In this article, we review the clinical trials of immune checkpoint inhibitors for advanced or recurrent endometrial cancer.
Collapse
Affiliation(s)
- Ami Jo
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Tadahiro Shoji
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan.
| | - Haruka Otsuka
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Marina Abe
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Shunsuke Tatsuki
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Yohei Chiba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Sho Sato
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Eriko Takatori
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Yoshitaka Kaido
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Takayuki Nagasawa
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Masahiro Kagabu
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, 2-1-1, Idaidori, Yahaba, Iwate, 028-3695, Japan
| |
Collapse
|
21
|
Song R, Jiang T, Zhang X, Shen C, Lou Q, Shan C. Triplet Electron Exchange in Carbon Nanodots-assisted Long-persistent near-infrared Chemiluminescence for Oncology Synergistic Imaging and Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411898. [PMID: 39661728 PMCID: PMC11791938 DOI: 10.1002/advs.202411898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/07/2024] [Indexed: 12/13/2024]
Abstract
In classical photodynamic therapy, tumor cells are killed by the cytotoxic species via type-I/II photochemical reactions, seriously limited by the external photoexcitation and hypoxia. Herein, the electron transfer mechanism between fluorophores and peroxalate-H2O2 reaction is investigated and the singlet/triplet electron exchange is utilized to achieve long-persistent chemiluminescence imaging and synergistic type-I/II/III photodynamic therapy. As a proof-of-concept, the photosensitizers of carbon nanodots (CDs)-loaded chlorin e6 (CDs-Ce6) are designed and integrated with the peroxalate molecules, and the as-prepare polymer carbon nanodots (p-CDs) exhibit novel tumor microenvironment (TME)-responsive long-persistent near-infrared CL and photochemical reactions, evoking the in vivo imaging and synergistic dynamic therapy in tumor tissue. Mechanistically, the excess reactive oxygen species in TME can trigger the chemically initiated singlet/triplet electron exchange between the hydrophobic CDs-Ce6 and peroxalate-derived 1,2-dioxetanes and thus the excess excited singlet/triplet electron of the CDs-Ce6 can ensure the long-persistent near-infrared CL, type I/II photochemical production of hydroxyl radicals, superoxide radical and singlet oxygen, and type III photochemical damage of maladjusted biomacromolecules, enabling the long-persistent near-infrared biological imaging and enhanced cancer therapy. These results shed a new sight into the energy transfer mechanism in chemiluminescence and pave a new sight into the architecture of multifunctional theragnostic nanoplatforms.
Collapse
Affiliation(s)
- Run‐Wei Song
- Henan Key Laboratory of Diamond Optoelectronic Materials and DevicesKey Laboratory of Material PhysicsMinistry of Educationand School of Physics and Laboratory of Zhongyuan LightZhengzhou UniversityZhengzhou450052China
| | - Tian‐Ci Jiang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450052China
| | - Xue‐Yang Zhang
- College of Public HealthZhengzhou UniversityZhengzhou450052China
| | - Cheng‐Long Shen
- Henan Key Laboratory of Diamond Optoelectronic Materials and DevicesKey Laboratory of Material PhysicsMinistry of Educationand School of Physics and Laboratory of Zhongyuan LightZhengzhou UniversityZhengzhou450052China
| | - Qing Lou
- Henan Key Laboratory of Diamond Optoelectronic Materials and DevicesKey Laboratory of Material PhysicsMinistry of Educationand School of Physics and Laboratory of Zhongyuan LightZhengzhou UniversityZhengzhou450052China
| | - Chong‐Xin Shan
- Henan Key Laboratory of Diamond Optoelectronic Materials and DevicesKey Laboratory of Material PhysicsMinistry of Educationand School of Physics and Laboratory of Zhongyuan LightZhengzhou UniversityZhengzhou450052China
| |
Collapse
|
22
|
Frumento D, Ţălu Ș. Light-based technologies in immunotherapy: advances, mechanisms and applications. Immunotherapy 2025; 17:123-131. [PMID: 40032620 PMCID: PMC11901425 DOI: 10.1080/1750743x.2025.2470111] [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: 10/16/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
Light-based immunotherapy uses specific wavelengths of light to activate or modulate immune responses. It primarily employs two mechanisms: direct activation of immune cells and indirect modulation of the tumor microenvironment (TME). Several light-based technologies are under investigation or clinical use in immunotherapy, including photodynamic immunotherapy (PDIT) and photothermal therapy (PTT). Optogenetic tools have the potential to precisely control T-cell receptor activation, cytokine release, or the activity of other immune effector cells. Light-based technologies present innovative opportunities within the realm of immunotherapy. The ability to precisely regulate immune cell activation via optogenetics, alongside the improved targeting of cancer cells through photoimmunotherapy, signifies a transformative shift in our strategies for immune modulation. Although many of these technologies remain in the experimental stage for various applications, initial findings are encouraging, especially concerning cancer treatment and immune modulation. Continued research and clinical trials are essential to fully harness the capabilities of light technology in the context of immune cell therapy.
Collapse
Affiliation(s)
| | - Ștefan Ţălu
- The Directorate of Research, Development and Innovation Management (DMCDI), The Technical University of Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
23
|
Garg P, Singhal G, Pareek S, Kulkarni P, Horne D, Nath A, Salgia R, Singhal SS. Unveiling the potential of gene editing techniques in revolutionizing Cancer treatment: A comprehensive overview. Biochim Biophys Acta Rev Cancer 2025; 1880:189233. [PMID: 39638158 DOI: 10.1016/j.bbcan.2024.189233] [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: 06/20/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Gene editing techniques have emerged as powerful tools in biomedical research, offering precise manipulation of genetic material with the potential to revolutionize cancer treatment strategies. This review provides a comprehensive overview of the current landscape of gene editing technologies, including CRISPR-Cas systems, base editing, prime editing, and synthetic gene circuits, highlighting their applications and potential in cancer therapy. It discusses the mechanisms, advantages, and limitations of each gene editing approach, emphasizing their transformative impact on targeting oncogenes, tumor suppressor genes, and drug resistance mechanisms in various cancer types. The review delves into population-level interventions and precision prevention strategies enabled by gene editing technologies, including gene drives, synthetic gene circuits, and precision prevention tools, for controlling cancer-causing genes, targeting pre-cancerous lesions, and implementing personalized preventive measures. Ethical considerations, regulatory challenges, and future directions in gene editing research for cancer treatment are also addressed. This review highlights how gene editing could revolutionize precision medicine by enhancing patient care and advancing cancer treatments with targeted, personalized methods. For these benefits to be fully realized, collaboration among researchers, doctors, regulators, and patient advocates is crucial in fighting cancer and meeting clinical needs.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Gargi Singhal
- Undergraduate Medical Sciences, S.N. Medical College Agra, Uttar Pradesh 282002, India
| | - Siddhika Pareek
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Aritro Nath
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S Singhal
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
24
|
Olawade DB, Clement David-Olawade A, Adereni T, Egbon E, Teke J, Boussios S. Integrating AI into Cancer Immunotherapy-A Narrative Review of Current Applications and Future Directions. Diseases 2025; 13:24. [PMID: 39851488 PMCID: PMC11764268 DOI: 10.3390/diseases13010024] [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: 12/20/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Cancer remains a leading cause of morbidity and mortality worldwide. Traditional treatments like chemotherapy and radiation often result in significant side effects and varied patient outcomes. Immunotherapy has emerged as a promising alternative, harnessing the immune system to target cancer cells. However, the complexity of immune responses and tumor heterogeneity challenges its effectiveness. OBJECTIVE This mini-narrative review explores the role of artificial intelligence [AI] in enhancing the efficacy of cancer immunotherapy, predicting patient responses, and discovering novel therapeutic targets. METHODS A comprehensive review of the literature was conducted, focusing on studies published between 2010 and 2024 that examined the application of AI in cancer immunotherapy. Databases such as PubMed, Google Scholar, and Web of Science were utilized, and articles were selected based on relevance to the topic. RESULTS AI has significantly contributed to identifying biomarkers that predict immunotherapy efficacy by analyzing genomic, transcriptomic, and proteomic data. It also optimizes combination therapies by predicting the most effective treatment protocols. AI-driven predictive models help assess patient response to immunotherapy, guiding clinical decision-making and minimizing side effects. Additionally, AI facilitates the discovery of novel therapeutic targets, such as neoantigens, enabling the development of personalized immunotherapies. CONCLUSIONS AI holds immense potential in transforming cancer immunotherapy. However, challenges related to data privacy, algorithm transparency, and clinical integration must be addressed. Overcoming these hurdles will likely make AI a central component of future cancer immunotherapy, offering more personalized and effective treatments.
Collapse
Affiliation(s)
- David B. Olawade
- Department of Allied and Public Health, School of Health, Sport and Bioscience, University of East London, London E16 2RD, UK
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK;
- Department of Public Health, York St John University, London E14 2BA, UK
| | | | - Temitope Adereni
- Department of Public Health, University of Dundee, Dundee DD1 4HN, UK;
| | - Eghosasere Egbon
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Life Science Engineering, FH Technikum, 1200 Vienna, Austria;
| | - Jennifer Teke
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK;
- Department of Surgery, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, Kent CT1 1QU, UK;
| | - Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, Kent CT1 1QU, UK;
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury, Kent CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK
| |
Collapse
|
25
|
Bhatnagar T, Haider M, Khan MY, Ashraf MZ. WGCNA and integrative network analysis identify CHRNA5 and CTLA4 as potential therapeutic targets against angiosarcoma. Cancer Treat Res Commun 2025; 42:100862. [PMID: 39832463 DOI: 10.1016/j.ctarc.2024.100862] [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: 07/02/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025]
Abstract
Angiosarcomas are a type of soft-tissue sarcoma characterized by aggressive malignant tumors originating from endothelial cells of blood vessels or lymphatic vessels. Limited studies have been done to explore the molecular pathophysiology of the disease, with rather limited studies involving transcriptomic analyzes. This study was undertaken to identify the shared molecular signatures and gene modules associated with angiosarcomas of various origin. Transcriptomic data analysis of publicly available data was done followed by WGCNA to identify shared signature gene modules. The Maximal Clique Centrality algorithm was applied to gene modules, and unclustered network analysis was conducted on differentially expressed genes to identify true hub genes. The expression of candidate genes in various cancer types was analyzed using GEPIA. WGCNA analysis identified five significant modules, with the most enriched module being associated with angiogenesis and cell junction regulators. The intersection of true hub genes from MCC analysis of WGCNA modules and high-degree nodes from an unclustered network revealed eight consistently overexpressed genes in all angiosarcoma samples.Among the eight enriched genes, CHRNA5 and CTLA4, are exclusively overexpressed in angiosarcoma and not in other cancers of the same tissue origin, with significant drug-protein interactions suggesting their potential as therapeutic targets.
Collapse
Affiliation(s)
- Trishla Bhatnagar
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India 110025
| | - Madiha Haider
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India 110025
| | - Mohd Yasir Khan
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India 110025
| | | |
Collapse
|
26
|
Hossain MA. A comprehensive review of targeting RAF kinase in cancer. Eur J Pharmacol 2025; 986:177142. [PMID: 39577552 DOI: 10.1016/j.ejphar.2024.177142] [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: 08/25/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024]
Abstract
RAF kinases, particularly the BRAF isoform, play a crucial role in the MAPK/ERK signaling pathway, regulating key cellular processes such as proliferation, differentiation, and survival. Dysregulation of this pathway often caused by mutations in the BRAF gene or alterations in upstream regulators like Ras and receptor tyrosine kinases contributes significantly to cancer development. Mutations, such as BRAF-V600E, are present in a variety of malignancies, with the highest prevalence in melanoma. Targeted therapies against RAF kinases have achieved substantial success, especially in BRAF-V600E-mutant melanomas, where inhibitors like vemurafenib and dabrafenib have demonstrated remarkable efficacy, leading to improved patient outcomes. These inhibitors have also shown clinical benefits in cancers such as thyroid and colorectal carcinoma, although to a lesser extent. Despite these successes, therapeutic resistance remains a major hurdle. Resistance mechanisms, including RAF dimerization, feedback reactivation of the MAPK pathway, and paradoxical activation of ERK signaling, often lead to diminished efficacy over time, resulting in disease progression or even secondary malignancies. In response, current research is focusing on novel therapeutic strategies, including combination therapies that target multiple components of the pathway simultaneously, such as MEK inhibitors used in tandem with RAF inhibitors. Additionally, next-generation RAF inhibitors are being developed to address resistance and enhance therapeutic specificity. This review discusses the clinical advancements in RAF-targeted therapies, with a focus on ongoing efforts to overcome therapeutic resistance and enhance outcomes for cancer patients. It also underscores the persistent challenges in effectively targeting RAF kinase in oncology.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
27
|
Zou H, Lai Y, Chen X, Ung COL, Hu H. Cost-effectiveness of camrelizumab plus rivoceranib versus sorafenib as first-line treatment of unresectable hepatocellular carcinoma. Therap Adv Gastroenterol 2025; 18:17562848241310314. [PMID: 39758963 PMCID: PMC11694292 DOI: 10.1177/17562848241310314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related mortality globally. Recent advancements in targeted therapies have improved outcomes for advanced HCC, yet therapeutic options remain limited. The CARES-310 trial demonstrated that camrelizumab plus rivoceranib significantly improves survival compared to sorafenib for advanced HCC. Objectives This study aimed to evaluate the cost-effectiveness of camrelizumab plus rivoceranib as a first-line treatment for unresectable HCC from the Chinese health system perspective. Design The cost-effectiveness analysis. Methods A partitioned survival model was constructed to estimate clinical and economic outcomes for patients with unresectable or metastatic HCC. The model included three health states: progression-free, progression disease, and death. The hypothetical cohort consisted of patients aged ⩾18 with HCC who had not received systemic therapy, reflecting the CARES-310 trial. Clinical data were derived from the CARES-310 trial and extrapolated using standard parameter distributions. Direct medical costs and utilities were sourced from the CARES-310 trial and published literature. Results The 10-year cost of camrelizumab plus rivoceranib was higher than sorafenib (USD 28,148.01 vs USD 20,997.86). Camrelizumab plus rivoceranib yielded an additional 0.26 quality-adjusted life-years (QALYs) with an incremental cost of USD 7150.15, resulting in an incremental cost-effectiveness ratio of USD 27,633.75/QALY. Sensitivity analyses confirmed the robustness of the base-case results. Conclusion Camrelizumab plus rivoceranib is likely a cost-effective first-line treatment for unresectable HCC from a Chinese health system perspective. This study highlights the need for additional real-world data to validate these findings and guide clinical decision-making for HCC.
Collapse
Affiliation(s)
- Huimin Zou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao
| | - Yunfeng Lai
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianwen Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao
| | - Carolina Oi Lam Ung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Taipa, Macao
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao
| | - Hao Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Taipa, Macao
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Taipa 999078, Macao
| |
Collapse
|
28
|
Tippareddy C, Martinez OM, Benza AR, Bera K, Ramaiya N, Tirumani SH. From guidelines to radiology practice: navigating the 2023 ASCO guidelines for advanced gastroesophageal cancer and beyond. Abdom Radiol (NY) 2025; 50:78-93. [PMID: 39123051 PMCID: PMC11711647 DOI: 10.1007/s00261-024-04499-y] [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: 04/07/2024] [Revised: 07/09/2024] [Accepted: 07/13/2024] [Indexed: 08/12/2024]
Abstract
The American Society of Clinical Oncology (ASCO) updated the guidelines for the treatment of advanced gastroesophageal (GE) cancer in 2023, signifying a major shift towards targeted therapeutics and precision medicine. This article serves as an imaging-based review of recent developments in the care of patients with GE cancer. We cover the epidemiology, the developing treatment paradigms, and the imaging assessment of GE malignancy. In addition, this review aims to familiarize radiologists with the unique adverse effects pertaining to therapeutics, surgeries, radiation therapies, and associated imaging corollaries. A case-based approach will be used to both explore the efficacy of modern treatments and demonstrate their adverse effects, such as chemotherapy-associated pneumonitis, radiation esophagitis, and anastomotic failure. With this comprehensive exploration of gastroesophageal cancer, radiologists will be equipped with the essential tools to inform the treatment decisions made by medical oncologists, radiation oncologists, and surgical oncologists in the new era of precision medicine.
Collapse
Affiliation(s)
- Charit Tippareddy
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA.
- Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | | | - Andrew R Benza
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kaustav Bera
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nikhil Ramaiya
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sree Harsha Tirumani
- Department of Radiology, University Hospitals Cleveland Medical Center, Case Western Reserve University, 1110 Euclid Ave, Cleveland, OH, 44106, USA
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
29
|
Patel MR, Falchook GS, Wang JS, Imedio ER, Kumar S, Miah K, Mugundu GM, Jones SF, Spigel DR, Hamilton EP. Open-Label, Multicenter, Phase I Study to Assess Safety and Tolerability of Adavosertib Plus Durvalumab in Patients with Advanced Solid Tumors. Target Oncol 2025; 20:127-138. [PMID: 39560862 PMCID: PMC11762568 DOI: 10.1007/s11523-024-01110-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Adavosertib (AZD1775) is a small-molecule Wee1 inhibitor. Durvalumab is a PD-L1 inhibitor. OBJECTIVE The safety, tolerability, pharmacokinetics, and preliminary antitumor activity of adavosertib plus durvalumab were evaluated in patients with refractory solid tumors to define the maximum tolerated dose (MTD) and recommended phase II dose (RP2D). PATIENTS AND METHODS This phase 1, non-randomized, open-label study determined MTD/RP2D using dose-escalation cohorts. Eligible patients had histologically confirmed tumors refractory to standard therapy or for which no standard of care existed. RESULTS A total of 55 patients received adavosertib with durvalumab. Overall, 3/52 evaluable patients experienced dose-limiting toxicities (DLTs; two grade 3 nausea, one grade 3 diarrhea that did not respond to care within 48 h). The most frequent (in > 5% of patients) treatment-emergent grade ≥ 3 toxicities were fatigue, diarrhea, nausea, anemia, and abdominal pain. MTD for twice-daily (bid) adavosertib dosing was oral adavosertib 150 mg bid (3 days on/4 days off; treatment days 15-17 and 22-24 of a 28-day cycle) with intravenous durvalumab 1500 mg four times a week (Q4W), which was also the RP2D. MTD for once-daily (qd) adavosertib dosing was oral adavosertib 300 mg qd (5 days on/2 days off; treatment days 15-19 and 22-26 of a 28-day cycle) with intravenous durvalumab 1500 mg Q4W. CONCLUSIONS This study defined the MTD/RP2D of adavosertib plus durvalumab in patients with advanced solid tumors. The safety profile of adavosertib with durvalumab was consistent with the known safety data of each agent. Findings provide preliminary evidence of limited antitumor activity of adavosertib plus durvalumab. TRIAL REGISTRATION ClinicalTrials.gov, NCT02617277 (registration: 30 November 2015).
Collapse
Affiliation(s)
- Manish R Patel
- Florida Cancer Specialists, 600 N Cattlemen Rd, Suite #200, Sarasota, FL, 34232, USA.
- Sarah Cannon Research Institute, Nashville, TN, USA.
| | | | - Judy S Wang
- Florida Cancer Specialists, 600 N Cattlemen Rd, Suite #200, Sarasota, FL, 34232, USA
- Sarah Cannon Research Institute, Nashville, TN, USA
| | | | | | - Kowser Miah
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Boston, MA, USA
| | - Ganesh M Mugundu
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Boston, MA, USA
| | | | | | | |
Collapse
|
30
|
Han J, Wang H. Immune Cell Homing Hydrogels for Cancer Immunotherapy. Methods Mol Biol 2025; 2902:107-116. [PMID: 40029598 DOI: 10.1007/978-1-0716-4402-7_6] [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: 03/05/2025]
Abstract
Cancer immunotherapy has shifted the paradigm for clinical cancer treatment in the past decade, especially with the success of checkpoint blockades and chimeric antigen receptor (CAR)-T cell therapy. However, the low patient response rate to checkpoint blockades, poor efficacy of CAR-T cell therapy against solid tumors, and severe side effects in both have limited the utility of cancer immunotherapy. These issues motivate the development of new immunotherapies that can induce persistent cytotoxic T lymphocyte response with minimal side effects. This often requires the targeted modulation of specific types of immune cells (e.g., dendritic cells and T cells) in lymphatic tissues or cancerous tissues, which is inevitably challenging. Immune cell homing materials, though, enable in situ recruitment and modulation of immune cells for the orchestration of systemic immune responses and overall antitumor efficacy. Here we introduce the design, synthesis, characterization, and immune analysis of dendritic cell-homing macroporous hydrogels for the development of cancer immunotherapy.
Collapse
Affiliation(s)
- Joonsu Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
31
|
Ma L, Kim MO. Advances in Preventive and Therapeutic Strategies for Oral Cancer: A Short Review. J Cancer Prev 2024; 29:113-119. [PMID: 39790224 PMCID: PMC11706729 DOI: 10.15430/jcp.24.027] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Oral cancer is a major global health concern, with high incidence and mortality rates, especially in high-risk populations. Early diagnosis remains a challenge, and current treatments, such as surgery, radiation, and chemotherapy, have limited effectiveness, particularly in advanced stages. Recent advances in targeted therapies and immunotherapy offer promising alternatives, providing more precise and personalized treatment options. Targeted therapies, such as epidermal growth factor receptor inhibitors, aim to disrupt specific molecular pathways in tumor growth, while immunotherapies, including immune checkpoint inhibitors and chimeric antigen receptor-T cell therapy, enhance the body's immune response to fight cancer. Combination therapies, integrating both targeted and immune strategies, are being explored to overcome the limitations of single-agent treatments. This review highlights the current strategies in the prevention and treatment of oral cancer, discusses emerging therapies, explores future research directions, focusing on optimizing existing treatments, identifying new biomarkers, and developing innovative therapeutic approaches. The potential of personalized medicine and combination therapies offers new hope for improving survival rates and quality of life for oral cancer patients.
Collapse
Affiliation(s)
- Lei Ma
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| |
Collapse
|
32
|
Kaviyarasan V, Das A, Deka D, Saha B, Banerjee A, Sharma NR, Duttaroy AK, Pathak S. Advancements in immunotherapy for colorectal cancer treatment: a comprehensive review of strategies, challenges, and future prospective. Int J Colorectal Dis 2024; 40:1. [PMID: 39731596 DOI: 10.1007/s00384-024-04790-w] [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] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Metastatic colorectal cancer (mCRC) continues to present significant challenges, particularly in patients with proficient mismatch repair/microsatellite stable (pMMR/MSS) tumors. This narrative review aims to provide recent developments in immunotherapy for CRC treatment, focusing on its efficacy and challenges. METHODS This review discussed the various immunotherapeutic strategies for CRC treatment, including immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1, combination therapies involving ICIs with other modalities, chimeric antigen receptor T-cell (CAR-T) cell therapy, and cancer vaccines. The role of the tumor microenvironment and immune evasion mechanisms was also explored to understand their impact on the effectiveness of these therapies. RESULTS This review provides a comprehensive update of recent advancements in immunotherapy for CRC, highlighting the potential of various immunotherapeutic approaches, including immune checkpoint inhibitors, combination therapies, CAR-T therapy, and vaccination strategies. The results of checkpoint inhibitors, particularly in patients with MSI-H/dMMR tumors, which have significant improvements in survival rates have been observed. Furthermore, this review also addresses the challenges faced in treating pMMR/MSS CRC, which remains resistant to immunotherapy. CONCLUSION Immunotherapy plays a significant role in the treatment of CRC, particularly in patients with MSI-H/dMMR tumors. However, many challenges remain, especially in treating pMMR/MSS CRC. This review discussed the need for further research into combination therapies, biomarker development, CAR-T cell therapy, and a deeper understanding of immune evasion mechanisms for CRC treatment.
Collapse
Affiliation(s)
- Vaishak Kaviyarasan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Alakesh Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Dikshita Deka
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Biki Saha
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| | - Neeta Raj Sharma
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| |
Collapse
|
33
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [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: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
34
|
Zhang J, Wei Q, Piao Y, Shao S, Zhou Z, Tang J, Xiang J, Shen Y. Synergistic Combination of Oral Transcytotic Nanomedicine and Histone Demethylase Inhibitor for Enhanced Cancer Chemoimmunotherapy. ACS NANO 2024; 18:33729-33742. [PMID: 39612220 DOI: 10.1021/acsnano.4c14816] [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: 11/30/2024]
Abstract
Oral nanomedicines present a preferable avenue for cancer immunotherapy, but their efficacy is limited by gastrointestinal absorption challenges, tumor physiopathologic barriers, and immune evasion mechanisms. Here, we present an approach that combines an oral transcytotic doxorubicin (DOX) nanomedicine with the histone demethylase inhibitor 5-carboxy-8-hydroxyquinoline (IOX1), thereby enabling synergistic chemoimmunotherapy. We demonstrate that IOX1 significantly augments the transcytosis capabilities of DOX-loaded poly(2-(N-oxide-N,N-diethylamino)ethylmethacrylate)-poly(ε-caprolactone) micelles (OPDOX), promoting their transcellular transport across various cellular barriers (villus, endothelial, and tumor cells), thus improving oral adsorption, vascular extravasation, and tumor penetration. Furthermore, IOX1 sensitizes chemotherapy to potentiate DOX-induced immunogenic cell death and downregulates programmed cell death-ligand 1 to disrupt the immune checkpoint mechanism, synergistically boosting robust antitumor immune responses. Consequently, orally administered OPDOX in combination with IOX1 efficiently inhibits CT26 tumor growth, highlighting the significant potential for enhancing the efficacy of oral nanomedicines in cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Jing Zhang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Qiuyu Wei
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
35
|
Zeng C, Ji J, Huang Y, Peng Y, Zhang X, Yang Z, Guo Z. A Novel Scoring System to Predict Acute Radiation Enteritis Recovery in Cervical Cancer Patients Undergoing Concurrent Chemoradiotherapy: A Southwest China Cohort Study. Int J Gen Med 2024; 17:5907-5919. [PMID: 39678675 PMCID: PMC11645290 DOI: 10.2147/ijgm.s485087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose To establish a pragmatic and effective predictive model for monitoring the recovery of radiation enteritis (RE) in cervical cancer patients undergoing concurrent chemoradiotherapy (CCRT). Methods This study included 105 cervical cancer patients undergoing CCRT. We assessed baseline clinicopathologic characteristics, evaluated the effects of CCRT on circulating immune cells, tumor biomarkers, and inflammatory cytokines, and developed a predictive scoring system, the Immune-Tumor-Score (ITS), using the LASSO-Cox regression model. The model performance of LASSO-Cox and nomogram was compared via ROC curve and calibration curve. Results The median age of the patients was 55 years, with 53.3% having a normal BMI and 46.7% having positive lymph nodes. Post-CCRT, significant decreases were observed in lymphocyte counts, T-cell subpopulations, and tumor markers (CA125, TPA, SCCA, CYFRA21). The CD4/CD8 ratio and IL10 levels were significantly higher post-CCRT, while inflammation indexes (NLR, ELR) increased, and LMR decreased. The ITS, derived from 11 significant parameters, effectively predicted RE recovery, outperforming a traditional nomogram. Higher ITS scores correlated with shorter RE recovery times, as validated by Kaplan-Meier analyses and ROC curves (AUC = 0.822). Conclusion The ITS system provides a robust and reliable tool for predicting RE recovery in cervical cancer patients undergoing CCRT, surpassing traditional models in accuracy and reliability. This tool enables better patient management by allowing for timely interventions and personalized treatment strategies. Future research should focus on validating these findings in larger cohorts and integrating additional clinical parameters to enhance the predictive power of the ITS.
Collapse
Affiliation(s)
- Chuan Zeng
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, People’s Republic of China
| | - Jia Ji
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, People’s Republic of China
| | - Yusheng Huang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, People’s Republic of China
| | - Yuan Peng
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, People’s Republic of China
| | - Xiaoyue Zhang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, People’s Republic of China
| | - Zhenzhou Yang
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, People’s Republic of China
| | - Zhengjun Guo
- Department of Cancer Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, People’s Republic of China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People’s Republic of China
| |
Collapse
|
36
|
Fei B, Yu M, Wang Z, Li S. Biomimetic Nanovaccines Restore Immunosuppressive Tumor Antigen-Presenting Cells via the Saposin-Feeding Strategy. ACS Biomater Sci Eng 2024; 10:7482-7491. [PMID: 39560472 DOI: 10.1021/acsbiomaterials.4c01337] [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: 11/20/2024]
Abstract
Cancer cell membrane-derived biomimetic nanovaccines have shown tremendous potential in cancer immunotherapy. However, their efficacy is restricted by the insufficient cross-presentation of cell membrane-associated antigens. Saposins (SAs), which are vital for membrane vesicle disintegration and cell membrane-associated antigen presentation, are severely deficient in the antigen-presenting cells (APCs) within tumors. Herein, we propose a complementary strategy for increasing the efficacy of biomimetic nanovaccines via the use of SAs. Biomimetic nanovaccines were designed using cancer cell membrane shells to provide a comprehensive array of tumor-associated antigens and reactive oxygen species (ROS)-responsive nanoparticle cores that allowed the codelivery of cytosine-guanine dinucleotides (CpGs) and SAs. The biomimetic nanovaccines were ROS-responsive and highly internalized by APCs, which enabled the release of CpGs and SAs in the endo/lysosomes of APCs. Furthermore, biomimetic nanovaccines increased the activation of immunosuppressive APCs and enhanced T-cell priming by delivering SAs to the APCs. Consequently, biomimetic nanovaccines loaded with SAs not only suppressed tumor growth but also exhibited excellent therapeutic effects in combination with immune checkpoint blockade strategies. Overall, our study provides insights into the development of enhanced biomimetic nanovaccines via integrating SAs and offers a promising strategy for highly effective cancer immunotherapy.
Collapse
Affiliation(s)
- Bingyuan Fei
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Miao Yu
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou 215123, China
| | - Shuo Li
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| |
Collapse
|
37
|
Zhang JY, Li XY, Li DX, Zhang ZH, Hu LQ, Sun CX, Zhang XN, Wu M, Liu LT. Endoplasmic reticulum stress in intestinal microecology: A controller of antineoplastic drug-related cardiovascular toxicity. Biomed Pharmacother 2024; 181:117720. [PMID: 39631125 DOI: 10.1016/j.biopha.2024.117720] [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: 08/16/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is extensively studied as a pivotal role in the pathological processes associated with intestinal microecology. In antineoplastic drug treatments, ER stress is implicated in altering the permeability of the mechanical barrier, depleting the chemical barrier, causing dysbiosis, exacerbating immune responses and inflammation in the immune barrier. Enteric dysbiosis and intestinal dysfunction significantly affect the circulatory system in various heart disorders. In antineoplastic drug-related cardiovascular (CV) toxicity, ER stress constitutes a web of relationships in the host-microbiome symbiotic regulatory loop. Therefore, understanding the holobiont perspective will help de-escalate spatial and temporal restrictions. This review investigates the role of ER stress-mediated gut microecological alterations in antineoplastic treatment-induced CV toxicity.
Collapse
Affiliation(s)
- Jing-Yi Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Ya Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - De-Xiu Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Zi-Hao Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lan-Qing Hu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Chang-Xin Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiao-Nan Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Long-Tao Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
38
|
Özalp FR, Yörükoğlu K, Yıldırım EÇ, Uzun M, Semiz HS. Prognostic value of B7-H3 expression in metastatic renal cell carcinoma and its impact on immunotherapy response. BMC Cancer 2024; 24:1471. [PMID: 39614178 DOI: 10.1186/s12885-024-13238-x] [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: 06/05/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is characterised by its immunogenic and proangiogenic nature and its resistance to conventional therapies. The advent of immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) has significantly improved patient survival, but resistance to these treatments remains a challenge. B7-H3, a potential immune checkpoint, has been implicated in modulating the tumour microenvironment and immune escape mechanisms in RCC. METHODS Immunohistochemical analysis of B7-H3 expression was performed in 84 metastatic RCC patients. Tissue microarrays and separate sections of formalin-fixed paraffin-embedded tissue were used for immunohistochemical staining. Membranous staining of the tumor cells was scored and statistical analyses were performed to assess the correlation between B7-H3 expression and treatment outcome. RESULTS B7-H3 expression was absent in 31% of patients, while 33.3% had a score of 1+, 31% had 2+, and 4.8% had 3+. High B7-H3 expression correlated with poorer OS (20 months vs. 45 months, p = 0.012). In patients receiving nivolumab, those with high B7-H3 expression had shorter PFS (2 months vs. 8 months, p = 0.037) and OS (17 months vs. 51 months, p = 0.01). B7-H3 expression was the only factor significantly affecting PFS and OS in multivariate analysis. CONCLUSION High B7-H3 expression is associated with poorer survival outcomes and reduced response to nivolumab in metastatic RCC patients. B7-H3 may serve as a predictive biomarker for immunotherapy response. Future studies should explore targeting B7-H3 in combination with existing therapies to enhance treatment efficacy.
Collapse
Affiliation(s)
- Faruk Recep Özalp
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey.
| | - Kutsal Yörükoğlu
- Department of Pathology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Eda Çalışkan Yıldırım
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Mehmet Uzun
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Hüseyin Salih Semiz
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
39
|
Yun D, Fagan E, Shin D, Back W, Lee S, Kim MS, Park H, Park JH, Kim YC. pH and Redox Dual-Responsive Nanoparticle with Enhanced Dendritic Cell Maturation for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64592-64608. [PMID: 39538128 DOI: 10.1021/acsami.4c15342] [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: 11/16/2024]
Abstract
Type I interferons (IFNs) are essential for activating dendritic cells (DCs) and presenting tumor-associated antigens to T cells. IFNs are primarily produced from DCs among immune cells. A combination of chemotherapy and metalloimmunotherapy induces IFN production by activating the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. However, chemotherapeutic agents deplete DC populations, suppressing immunostimulatory activities, despite their potent anticancer activities. Furthermore, an optimal ratio between chemotherapeutic agents and metal for activating DCs at the highest level has not been reported, and evidence for ensuring DC survival is lacking. In this study, we hypothesized that there is an optimal ratio to yield the highest DC maturation and anticancer activity with minimal DC depletion. To demonstrate it, we have designed a pH and redox dual-responsive nanoparticle, MnO2@BSA@DOX (MD), to prevent DCs from depleting and activate the cGAS-STING pathway both in cancer cells and DCs, inducing considerable levels of IFNs and maturation. MD consists of a core-layer structure, a manganese dioxide (MnO2) core, and a cross-linked layer with bovine serum albumin (BSA) and doxorubicin (DOX), with a specific ratio of DOX to manganese. MD exhibits structure-based selectivity between cancer cells and DCs by targeting the extracellular pH of the tumor microenvironment and intracellular redox reactions in cancer cells. Among various formulations, the 1:1 ratio shows the highest maturation with no significant depletion. Moreover, it induces distinct cytotoxicity in cancer cells through apoptosis and cGAS-STING activation, leading to increased calreticulin expression and enhanced DC phagocytosis. Consequently, it results in superior tumor suppression and prolonged survival with the high accumulation of MD in the tumor and no observed systemic toxicities, highlighting its potential as a therapeutic agent in cancer treatments.
Collapse
Affiliation(s)
- Dohyun Yun
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Erinn Fagan
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Dongik Shin
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Woojin Back
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Susam Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Mun Sik Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Heewon Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| |
Collapse
|
40
|
Gharib E. Closing Editorial: Colorectal Cancer-A Molecular Genetics Perspective. Int J Mol Sci 2024; 25:12604. [PMID: 39684316 DOI: 10.3390/ijms252312604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Colorectal cancer (CRC) remains a significant global health challenge, ranking third in incidence and second in mortality among all cancers [...].
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
41
|
Silva MLS. Lectin-modified drug delivery systems - Recent applications in the oncology field. Int J Pharm 2024; 665:124685. [PMID: 39260750 DOI: 10.1016/j.ijpharm.2024.124685] [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: 03/01/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Chemotherapy with cytotoxic drugs remains the core treatment for cancer but, due to the difficulty to find general and usable biochemical differences between cancer cells and normal cells, many of these drugs are associated with lack of specificity, resulting in side effects and collateral cytotoxicity that impair patients' adherence to therapy. Novel cancer treatments in which the cytotoxic effect is maximized while adverse effects are reduced can be implemented by developing targeted therapies that exploit the specific features of cancer cells, such as the typical expression of aberrant glycans. Modification of drug delivery systems with lectins is one of the strategies to implement targeted chemotherapies, as lectins are able to specifically recognize and bind to cancer-associated glycans expressed at the surface of cancer cells, guiding the drug treatment towards these cells and not affecting healthy ones. In this paper, recent advances on the development of lectin-modified drug delivery systems for targeted cancer treatments are thoroughly reviewed, with a focus on their properties and performance in diverse applications, as well as their main advantages and limitations. The synthesis and analytical characterization of the cited lectin-modified drug delivery systems is also briefly described. A comparison with free-drug treatments and with antibody-modified drug delivery systems is presented, emphasizing the advantages of lectin-modified drug delivery systems. Main constraints and potential challenges of lectin-modified drug delivery systems, including key difficulties for clinical translation of these systems, and the required developments in this area, are also signalled.
Collapse
Affiliation(s)
- Maria Luísa S Silva
- Centro de Estudos Globais, Universidade Aberta, Rua da Escola Politécnica 147, 1269-001 Lisboa, Portugal.
| |
Collapse
|
42
|
Li Z, Zhou Z, Zhang N, Tian B, Chen X, Zhao H, Wang H. Hepatitis associated with immune checkpoint inhibitors-based combinations of other therapies: A real-world pharmacovigilance analysis based on the FDA adverse event reporting system (FAERS) database. Cancer Immunol Immunother 2024; 74:25. [PMID: 39546001 PMCID: PMC11568091 DOI: 10.1007/s00262-024-03858-4] [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: 03/10/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND The combination regimen with immune checkpoint inhibitors (ICIs) and other therapies has been widely applied for patients with non-small-cell lung cancer (NSCLC). To date, no literature has systematically addressed the risk of hepatitis associated with the combination therapy. We conducted this pharmacovigilance analysis using the Food and Drug Administration Adverse Event Reporting System (FAERS). PATIENTS AND METHODS A total of 587,016 NSCLC reports were extracted from FAERS database spanning from the first quarter of 2013 to the second quarter of 2023. After filtering duplicate reports, logistic regression model was used to detect safety signals, and multivariable logistic regression model was used to confirm the interaction between ICI and other drugs. RESULTS Of the 81,512 patients with NSCLC, 2785 cases developed hepatitis. Multivariable logistic regression analyses revealed that the adjusted ROR of ICI combined with targeted therapy (TT) was the highest 1.64 (95% CI, 1.32-2.02; P < 0.0001) among all therapies, while that of TT and ICI treatment were 1.07 (95% CI, 0.98-1.17; P = 0.1097) and 1.12 (95% CI, 1.03-1.22; P = 0.0111), respectively. The adjusted ROR for the interaction effect was 1.64 (95% CI, 1.32-2.02; P < 0.0001). Furthermore, the adjusted ROR of ICI combined with KRAS-targeted drugs was the highest 3.03 (95% CI, 1.49-5.93; P = 0.0016) among all targeted drugs, with an adjusted ROR of 3.03 (95% CI, 1.49-5.93; P = 0.0016), indicating a meaningful interaction of these two kinds of drugs. CONCLUSION We confirmed that combination treatment of ICI and TT is associated with the amplified risk of hepatitis, which is partly due to the interaction between ICI and TT, and the KRAS-targeted drugs may harbor the highest potential for hepatitis induction among TT drugs when combined with ICI. Besides, the combination treatment of ICI- and KRAS-targeted drugs also increases the incidence of colitis, pulmonary embolism and dehydration.
Collapse
Affiliation(s)
- Zhaohui Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Zixiang Zhou
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- Peking Union Medical College, Beijing, China
| | - Nan Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Binhe Tian
- Division of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Xiangqi Chen
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
- Peking Union Medical College, Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Hanping Wang
- Division of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Beijing, 100730, China.
| |
Collapse
|
43
|
Xie L, Dai X, Li Y, Cao Y, Shi M, Li X. Pickering Emulsion of Curcumin Stabilized by Cellulose Nanocrystals/Chitosan Oligosaccharide: Effect in Promoting Wound Healing. Pharmaceutics 2024; 16:1411. [PMID: 39598536 PMCID: PMC11597753 DOI: 10.3390/pharmaceutics16111411] [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: 05/20/2024] [Revised: 08/20/2024] [Accepted: 09/27/2024] [Indexed: 11/29/2024] Open
Abstract
Background: The stabilization of droplets in Pickering emulsions using solid particles has garnered significant attention through various methods. Cellulose and chitin derivatives in nature offer a sustainable source of Pickering emulsion stabilizers. Methods: In this study, medium-chain triglycerides were used as the oil phase for the preparation of emulsion. This study explores the potential of cellulose nanocrystals (CNC) and shell oligosaccharides (COS) as effective stabilizers for achieving stable Pickering emulsions. Optical microscopy, CLSM, and Cyro-SEM were employed to analyze CNC/COS-Cur, revealing the formation of bright and uniform yellow spherical emulsions. Results: CLSM and SEM results confirmed that CNC/COS formed a continuous and compact shell at the oil-water interface layer, enabling a stable 2~3 microns Pickering emulsion with CNS/COS-Cur as an oil-in-water emulsion stabilizer. Based on FTIR, XRD, and SEM analyses of CNC/COS, along with zeta potential measurements of the emulsion, we found that CNC and COS complexed via electrostatic adsorption, forming irregular rods measuring approximately 200-300 nm in length. An evaluation of the DPPH radical-scavenging ability demonstrated that the CNC/ COS-Cur Pickering emulsion performed well in vitro. In vivo experiments involving full-thickness skin excision surgery in rats revealed that CNC/COS-Cur facilitated wound repair processes. Measurements of the MDA and SOD content in healing tissues indicated that the CNC/COS-Cur Pickering emulsion increased SOD levels and reduced MDA content, effectively countering oxidative stress-induced damage. An assessment based on wound-healing rates and histopathological examination showed that CNC/COS-Cur promoted granulation tissue formation, fibroblast proliferation, angiogenesis, and an accelerated re-epithelialization process within the wound tissue, leading to enhanced collagen deposition and facilitating rapid wound-healing capabilities. An antibacterial efficacy assessment conducted in vitro demonstrated antibacterial activity.
Collapse
Affiliation(s)
- Long Xie
- Science and Education Section, The First People’s Hospital of Shuangliu District, Chengdu (West China Airport Hospital Sichuan University), Chengdu 610299, China;
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.D.); (Y.L.); (Y.C.)
| | - Xiaolin Dai
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.D.); (Y.L.); (Y.C.)
- Department of pharmacy, Chengdu Seventh People’s Hospital (Affliated Cancer Hospital of Chengdu Medical College), Chengdu 610203, China
| | - Yuke Li
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.D.); (Y.L.); (Y.C.)
| | - Yi Cao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.D.); (Y.L.); (Y.C.)
| | - Mingyi Shi
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Xiaofang Li
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (X.D.); (Y.L.); (Y.C.)
| |
Collapse
|
44
|
Song D, Hou S, Ma N, Yan B, Gao J. Efficacy and safety of PD-1/PD-L1 and CTLA-4 immune checkpoint inhibitors in the treatment of advanced colorectal cancer: a systematic review and meta-analysis. Front Immunol 2024; 15:1485303. [PMID: 39555073 PMCID: PMC11563947 DOI: 10.3389/fimmu.2024.1485303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Background The efficacy and safety of PD-1/PD-L1 inhibitors combined with CTLA-4 inhibitors in the treatment of advanced colorectal cancer is controversial. This meta-analysis aimed to evaluate the efficacy and safety of PD-1/PD-L1 inhibitors combined with CTLA-4 inhibitors for advanced colorectal cancer. Methods PubMed, Embase, the Cochrane Library, and Web of Science databases were systematically searched for relevant studies. Outcomes including median progression-free survival (mPFS), median overall survival (mOS), overall response rate (ORR), disease control rate (DCR), treatment-related adverse events (TRAEs) and ≥grade 3 TRAEs were extracted for further analysis. The risk of bias was assessed by subgroup analysis. Results 12 articles with 566 patients were identified and subjected to meta-analysis. With regard to survival analysis, the pooled mOS and mPFS were 6.66 months (95%CI 4.85-9.16) and 2.92 months (95%CI 2.23-3.83), respectively. In terms of tumor response, the pooled ORR and DCR were 21% (95%CI 6%-41%) and 49% (95%CI 27%-71%), respectively. The pooled AEs rate and ≥ grade 3 AEs rate were 94% (95%CI 86%-99%) and 44% (95%CI 30%-58%). Conclusion PD-1/PD-L1 inhibitors combined with CTLA-4 inhibitors have shown promising clinical responses in the treatment of colorectal cancer (CRC). Although the incidence of adverse reactions is high, they are generally tolerable. Systematic review registration https://inplasy.com/, identifier INPLASY202480030.
Collapse
Affiliation(s)
- Dandan Song
- Department of Neurology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shufu Hou
- Department of Gastrointestinal Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ning Ma
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Bing Yan
- Department of Gastrointestinal Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Gao
- Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
45
|
Song Q, Xu Y, Zhang M, Wu L, Liu S, Lv Y, Hu T, Zhao J, Zhang X, Xu X, Li Q, Zhou M, Zhang X, Lu P, Yu G, Zhao C, Yang J. A β-1,3/1,6-glucan enhances anti-tumor effects of PD1 antibody by reprogramming tumor microenvironment. Int J Biol Macromol 2024; 279:134660. [PMID: 39134196 DOI: 10.1016/j.ijbiomac.2024.134660] [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: 04/21/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024]
Abstract
Checkpoint blockades have emerged as a frontline approach in cancer management, designed to enhance the adaptive immune response against tumors. However, its clinical efficacy is limited to a narrow range of tumor types, which necessitates the exploration of novel strategies that target another main branch of the immune system. One such potential strategy is the therapeutic modulation of pattern recognition receptors (PRRs) pathways in innate immune cells, which have shown promise in tumor eradication. Previously, a β-1,3/1,6-glucan with high purity from Durvillaea antarctica (BG136) was reported by our group to exhibit pan-antitumor effects. In the current study, we systemically studied the antitumor activity of BG136 in combination with anti-PD1 antibody in MC38 syngeneic tumor model in vivo. Integrated transcriptomic and metabolomic analyses suggested that BG136 enhanced the antitumor immunity of anti-PD1 antibody by reprogramming the tumor microenvironment to become more proinflammatory. In addition, an increase in innate and adaptive immune cell infiltration and activation, enhanced lipid metabolism, and a decrease in ascorbate and aldarate metabolism were also found. These findings provide mechanistic insights that support the potent antitumor efficacy of BG136 when combined with immune checkpoint inhibitor antibodies.
Collapse
Affiliation(s)
- Qiaoling Song
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Yuting Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Minghui Zhang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Lijuan Wu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Shan Liu
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Youjing Lv
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Ting Hu
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Jun Zhao
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Xiaonan Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Xiaohan Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Quancai Li
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Mingming Zhou
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Xinxin Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China; Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Peizhe Lu
- Department of Neuroscience, University of Michigan, Ann Arbor, MI 48103, USA
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Laoshan Laboratory, Qingdao 266237, China
| | - Chenyang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China.
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Marine Science and Technology Center, Qingdao 266100, China.
| |
Collapse
|
46
|
Garg P, Pareek S, Kulkarni P, Horne D, Salgia R, Singhal SS. Next-Generation Immunotherapy: Advancing Clinical Applications in Cancer Treatment. J Clin Med 2024; 13:6537. [PMID: 39518676 PMCID: PMC11546714 DOI: 10.3390/jcm13216537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Next-generation immunotherapies have revolutionized cancer treatment, offering hope for patients with hard-to-treat tumors. This review focuses on the clinical applications and advancements of key immune-based therapies, including immune checkpoint inhibitors, CAR-T cell therapy, and new cancer vaccines designed to harness the immune system to combat malignancies. A prime example is the success of pembrolizumab in the treatment of advanced melanoma, underscoring the transformative impact of these therapies. Combination treatments, integrating immunotherapy with chemotherapy, radiation, and targeted therapies, are demonstrating synergistic benefits and improving patient outcomes. This review also explores the evolving role of personalized immunotherapy, guided by biomarkers, genomic data, and the tumor environment, to better target individual tumors. Although significant progress has been made, challenges such as resistance, side effects, and high treatment costs persist. Technological innovations, including nanotechnology and artificial intelligence, are explored as future enablers of these therapies. The review evaluates key clinical trials, breakthroughs, and the emerging immune-modulating agents and advanced delivery systems that hold great promise for enhancing treatment efficacy, reducing toxicity, and expanding access to immunotherapy. In conclusion, this review highlights the ongoing advancements in immunotherapy that are reshaping cancer care, with future strategies poised to overcome current challenges and further extend therapeutic reach.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Siddhika Pareek
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Departments of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
47
|
Cen S, Yuan M, Sun Q, Hou G, Ying J, Xu Q, Zheng Y, Dong Y, Pan H, Han W. Efficacy and safety of dual blockade of HER2 and PD-1 in patients with HER2-positive gastric cancer: a retrospective, multicentre study. Sci Rep 2024; 14:25030. [PMID: 39443515 PMCID: PMC11500361 DOI: 10.1038/s41598-024-76296-y] [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: 06/12/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) expression is one of the most important pathological characteristics of gastric cancer. The positive rate of HER2 expression in patients with gastric cancer is approximately 20%. The phase III Keynote-811 study revealed that anti-HER2 and anti-PD-1 therapy combined with chemotherapy could significantly improve the objective response rate as first-line treatment in patients with HER2-positive advanced gastric cancer. In the present study, we aimed to evaluate the efficacy of combination therapy with trastuzumab and PD-1 inhibitors in patients with advanced HER2-positive gastric cancer in a real-world setting. Seventy-two HER2-positive gastric cancer patients from three hospitals in China were retrospectively reviewed. These patients were treated with trastuzumab plus one anti-PD-1 agent with or without chemotherapy. The overall response rate, progression-free survival and overall survival were assessed according to the Response Evaluation Criteria in Solid Tumours (RECIST 1.1). From January 2018 to October 2021, 72 patients with HER2-positive gastric cancer received trastuzumab and a PD-1 inhibitor with or without chemotherapy as neoadjuvant chemotherapy, first-line therapy, second-line therapy or salvage therapy. The ORR was 54.2% for all patients and 79.4% for previously untreated patients. The median PFS and median OS were 10 months (95% CI: 8-13 months) and 26.1 months (95% CI: 18.5-NA months), respectively, for all patients. Grade 3 adverse effects occurred in approximately 25% of patients. Immune-related adverse effects occurred in approximately 12.5% of patients. Trastuzumab and PD-1 inhibitor combination therapy with or without chemotherapy achieved satisfactory survival outcomes in patients with HER2-positive gastric cancer with acceptable safety.
Collapse
Affiliation(s)
- Shuyi Cen
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China
| | - Meiqin Yuan
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Qunan Sun
- Department of Medical Oncology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, PRC, China
| | - Guilan Hou
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Jieer Ying
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Qi Xu
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China
| | - Yu Zheng
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China
| | - Ying Dong
- Department of Medical Oncology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, 88# Jiefang Road, Hangzhou, 310009, Zhejiang, PRC, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China.
| | - Weidong Han
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine(HIM), Chinese Academy of Sciences, 38# Guangji Road, Hangzhou, 310022, Zhejiang, PRC, China.
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3# East Qinchun Road, Hangzhou, 310016, Zhejiang, PRC, China.
| |
Collapse
|
48
|
Tang L, Tian H, Min Q, You H, Yin M, Yang L, Zhao Y, Wu X, Li M, Du F, Chen Y, Deng S, Li X, Chen M, Gu L, Sun Y, Xiao Z, Li W, Shen J. Decoding the epitranscriptome: a new frontier for cancer therapy and drug resistance. Cell Commun Signal 2024; 22:513. [PMID: 39434167 PMCID: PMC11492518 DOI: 10.1186/s12964-024-01854-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: 07/04/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024] Open
Abstract
As the role of RNA modification in gene expression regulation and human diseases, the "epitranscriptome" has been shown to be an important player in regulating many physiological and pathological processes. Meanwhile, the phenomenon of cancer drug resistance is becoming more and more frequent, especially in the case of cancer chemotherapy resistance. In recent years, research on relationship between post-transcriptional modification and cancer including drug resistance has become a hot topic, especially the methylation of the sixth nitrogen site of RNA adenosine-m6A (N6-methyladenosine). m6A modification is the most common post-transcriptional modification of eukaryotic mRNA, accounting for 80% of RNA methylation modifications. At the same time, several other modifications of RNA, such as N1-methyladenosine (m1A), 5-methylcytosine (m5C), 3-methylcytosine (m3C), pseudouridine (Ψ) and N7-methylguanosine (m7G) have also been demonstrated to be involved in cancer and drug resistance. This review mainly discusses the research progress of RNA modifications in the field of cancer and drug resistance and targeting of m6A regulators by small molecule modulators, providing reference for future study and development of combination therapy to reverse cancer drug resistance.
Collapse
Affiliation(s)
- Lu Tang
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Scientific Research and Experimental Training Center, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| | - Hua Tian
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qi Min
- Department of Pharmacy, Mianyang Hospital of TCM, Sichuan Mianyang, 621000, China
| | - Huili You
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Mengshuang Yin
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Liqiong Yang
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Yueshui Zhao
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Xu Wu
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Mingxing Li
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Fukuan Du
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Yu Chen
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Shuai Deng
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Xiaobing Li
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Meijuan Chen
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Li Gu
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yuhong Sun
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zhangang Xiao
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China.
| | - Wanping Li
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China.
| | - Jing Shen
- Department of Pharmacology, School of Pharmacy, Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, Sichuan, 646000, China.
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China.
| |
Collapse
|
49
|
Guo Y, Patel HJ, Patel AS, Squillante E, Patel K. Albendazole nanosuspension coated granules for the rapid localized release and treatment of colorectal cancer. Colloids Surf B Biointerfaces 2024; 245:114320. [PMID: 39423765 DOI: 10.1016/j.colsurfb.2024.114320] [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: 06/17/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/21/2024]
Abstract
Albendazole (ABZ), an anthelmintic drug, has been repurposed to treat various types of cancers. However, poor solubility of ABZ, resulting in low bioavailability, limits its application. Nanosuspension is a versatile method for enhancing the dissolution of hydrophobic molecules, but a successful drying has been the biggest challenge in the field. The objective of this research is to formulate and optimize ABZ nanosuspension (NS) coated granules for rapid delivery of ABZ for the treatment of colorectal cancer. ABZ NS was prepared by dual centrifugation method using Kollidon® VA64 and sodium lauryl sulphate (SLS) as stabilizers. The processing method was optimized to obtain a stable nanosuspension with particle size < 300 nm. The optimized ABZ NS was coated on microcrystalline cellulose (MCC) to form the nano-coated granules (NCG) and filled in EUDRACAP® for colon targeted delivery. The ABZ NS and NCG achieved ∼ 60 % and ∼55 % drug release, respectively, in presence of bile salt at colonic pH. Half-maximal inhibitory concentration (IC50) of ABZ NS was found to be 1.18 ± 0.081 µM and 3.59 ± 0.080 µM in two colorectal cancer cell lines: HCT 116 and HT-29, respectively. In addition, In vitro 3D tumor assay revealed that ABZ NS has superior tumor growth inhibition activity compared to the control and pure ABZ. The preparation of ABZ NCG in EUDRACAP® could be a promising approach to achieve colon targeted delivery and to repurpose ABZ for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Yi Guo
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Henis J Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Akanksha S Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA
| | - Emilio Squillante
- College of Pharmacy and Health Sciences, St. John's University, NY, USA.
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, NY, USA.
| |
Collapse
|
50
|
Sancho-Albero M, Fenaroli AL, Scaccaglia M, Matteo C, Grasselli C, Zucchetti M, Frapolli R, Nastasi C, De Cola L. Two Different Responsive Organosilica Nanocarriers to Combine Chemo- and Immunotherapy against Cancer. ACS OMEGA 2024; 9:41225-41235. [PMID: 39398182 PMCID: PMC11465578 DOI: 10.1021/acsomega.4c02838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/22/2024] [Accepted: 08/08/2024] [Indexed: 10/15/2024]
Abstract
The combination of chemo- and immunotherapy was recently demonstrated to improve a patient's response to therapy, giving rise to an emerging cancer treatment known as chemoimmunotherapy (CIT). Despite the promising benefits of CIT, the most important challenges are (i) the simultaneous or time-controlled delivery of two drugs and (ii) the selective uptake into different cells for each of the drugs: cancer cells for the chemotherapeutic and macrophages for the immunostimulation actives. Herein, a delivery strategy based on morphologically different stimuli-responsive breakable organosilica nanocarriers is exploited to transport two distinct drugs in the different cells using different times of delivery. We employ stimulus-sensitive, PEGylated organosilica nanocages to encapsulate the chemotherapeutic agent doxorubicin, which is preferentially taken up by tumor cells vs macrophages. On the other hand, similar size mesoporous organosilica nanoparticles, preferentially internalized by macrophages, are filled with the immunostimulator resiquimod. The administration in a sequential manner of the two different nanocarriers allowed us to assess the integrated effect of the combined therapy versus treatment with a single drug. In vitro work clearly shows an important reduction of tumor cell viability when both chemo- and immunotherapeutic agents are delivered.
Collapse
Affiliation(s)
- Maria Sancho-Albero
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Alessia Lucrezia Fenaroli
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Mirco Scaccaglia
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Cristina Matteo
- Department
of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Chiara Grasselli
- Department
of Oncology, Immunopharmacology Unit, Istituto
di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Massimo Zucchetti
- Department
of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Roberta Frapolli
- Department
of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Claudia Nastasi
- Department
of Oncology, Immunopharmacology Unit, Istituto
di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
| | - Luisa De Cola
- Department
of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan 20156, Italy
- Department
of Pharmaceutical Science, DISFARM, Università
degli Studi di Milano, Milan 20133, Italy
| |
Collapse
|